101
|
Vilar R, Fish RJ, Casini A, Neerman-Arbez M. Fibrin(ogen) in human disease: both friend and foe. Haematologica 2020; 105:284-296. [PMID: 31949010 PMCID: PMC7012490 DOI: 10.3324/haematol.2019.236901] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 11/21/2019] [Indexed: 12/19/2022] Open
Abstract
Fibrinogen is an abundant protein synthesized in the liver, present in human blood plasma at concentrations ranging from 1.5-4 g/L in healthy individuals with a normal half-life of 3-5 days. With fibrin, produced by thrombin-mediated cleavage, fibrinogen plays important roles in many physiological processes. Indeed, the formation of a stable blood clot, containing polymerized and cross-linked fibrin, is crucial to prevent blood loss and drive wound healing upon vascular injury. A balance between clotting, notably the conversion of fibrinogen to fibrin, and fibrinolysis, the proteolytic degradation of the fibrin mesh, is essential. Disruption of this equilibrium can cause disease in distinct manners. While some pathological conditions are the consequence of altered levels of fibrinogen, others are related to structural properties of the molecule. The source of fibrinogen expression and the localization of fibrin(ogen) protein also have clinical implications. Low levels of fibrinogen expression have been detected in extra-hepatic tissues, including carcinomas, potentially contributing to disease. Fibrin(ogen) deposits at aberrant sites including the central nervous system or kidney, can also be pathological. In this review, we discuss disorders in which fibrinogen and fibrin are implicated, highlighting mechanisms that may contribute to disease.
Collapse
Affiliation(s)
- Rui Vilar
- Department of Genetic Medicine and Development, University of Geneva Faculty of Medicine
| | - Richard J Fish
- Department of Genetic Medicine and Development, University of Geneva Faculty of Medicine
| | - Alessandro Casini
- Division of Angiology and Hemostasis, University Hospitals and University of Geneva Faculty of Medicine
| | - Marguerite Neerman-Arbez
- Department of Genetic Medicine and Development, University of Geneva Faculty of Medicine .,iGE3, Institute of Genetics and Genomics in Geneva, Geneva, Switzerland
| |
Collapse
|
102
|
Kapnisis K, Seidner H, Prokopi M, Pasias D, Pitsillides C, Anayiotos A, Kaliviotis E. The effects of stenting on hemorheological parameters: An in vitro investigation under various blood flow conditions. Clin Hemorheol Microcirc 2019; 72:375-393. [PMID: 31006672 PMCID: PMC7739967 DOI: 10.3233/ch-180540] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Despite their wide clinical usage, stent functionality may be compromised by complications at the site of implantation, including early/late stent thrombosis and occlusion. Although several studies have described the effect of fluid-structure interaction on local haemodynamics, there is yet limited information on the effect of the stent presence on specific hemorheological parameters. The current work investigates the red blood cell (RBC) mechanical behavior and physiological changes as a result of flow through stented vessels. Blood samples from healthy volunteers were prepared as RBC suspensions in plasma and in phosphate buffer saline at 45% haematocrit. Self-expanding nitinol stents were inserted in clear perfluoroalkoxy alkane tubing which was connected to a syringe, and integrated in a syringe pump. The samples were tested at flow rates of 17.5, 35 and 70 ml/min, and control tests were performed in non-stented vessels. For each flow rate, the sample viscosity, RBC aggregation and deformability, and RBC lysis were estimated. The results indicate that the presence of a stent in a vessel has an influence on the hemorheological characteristics of blood. The viscosity of all samples increases slightly with the increase of the flow rate and exposure. RBC aggregation and elongation index (EI) decrease as the flow rate and exposure increases. RBC lysis for the extreme cases is evident. The results indicate that the stresses developed in the stent area for the extreme conditions could be sufficiently high to influence the integrity of the RBC membrane.
Collapse
Affiliation(s)
- K Kapnisis
- Department of Mechanical Engineering and Material Science and Engineering, Cyprus University of Technology, Limasol, Cyprus
| | - H Seidner
- Department of Mechanical Engineering and Material Science and Engineering, Cyprus University of Technology, Limasol, Cyprus
| | - M Prokopi
- Department of Mechanical Engineering and Material Science and Engineering, Cyprus University of Technology, Limasol, Cyprus
| | - D Pasias
- Department of Mechanical Engineering and Material Science and Engineering, Cyprus University of Technology, Limasol, Cyprus
| | - C Pitsillides
- Department of Mechanical Engineering and Material Science and Engineering, Cyprus University of Technology, Limasol, Cyprus
| | - A Anayiotos
- Department of Mechanical Engineering and Material Science and Engineering, Cyprus University of Technology, Limasol, Cyprus
| | - E Kaliviotis
- Department of Mechanical Engineering and Material Science and Engineering, Cyprus University of Technology, Limasol, Cyprus.,Department of Mechanical Engineering, University College London, London, UK
| |
Collapse
|
103
|
Abstract
OBJECTIVE Individuals with HIV suffer a higher burden of cardiovascular diseases. Traditional cardiovascular risk scores consistently underestimate cardiovascular risk in this population. Subsets of microRNAs (miRNAs) are differentially expressed among individuals with cardiovascular disease and individuals infected with HIV. However, no study has clarified whether specific miRNAs may be biomarkers for cardiovascular disease in individuals with HIV. DESIGN/METHODS We compared the miRNA expression profiles of 34 HIV-positive individuals who had experienced clinically adjudicated type I myocardial infarctions (MI) with the profiles of 76 HIV-positive controls matched by traditional cardiovascular risk factors and HIV-specific measures. Using the elastic net algorithm, we selected miRNAs most strongly associated with incident MI and then used conditional Cox proportional hazards regression and cross-validation to evaluate miRNAs and their association with incident MI. We evaluated whether miRNA markers would improve risk classification relative to the Framingham Risk Score. RESULTS Higher miR-125a-5p and miR-139-5p expression levels were each associated with increased risk of developing MI after adjustment for Framingham Risk Score and HIV-related factors (hazard ratio 2.43, P = 0.018; hazard ratio 2.13, P = 0.048, respectively). Compared with the Framingham Risk Score alone, adding expression levels of miR-125a-5p or miR-139-5p resulted in an integrated discrimination improvement of 10.1 or 5.8%, respectively. CONCLUSION MiR-125a-5p and miR-139-5p, transcripts known to be differentially expressed in HIV-positive individuals, may serve as unique biomarkers predictive of cardiovascular disease in these patients and may help clarify processes because of HIV infection that contribute to cardiovascular disorders in this population.
Collapse
|
104
|
Ohtsuki T, Satoh K, Shimizu T, Ikeda S, Kikuchi N, Satoh T, Kurosawa R, Nogi M, Sunamura S, Yaoita N, Omura J, Aoki T, Tatebe S, Sugimura K, Takahashi J, Miyata S, Shimokawa H. Identification of Adipsin as a Novel Prognostic Biomarker in Patients With Coronary Artery Disease. J Am Heart Assoc 2019; 8:e013716. [PMID: 31752640 PMCID: PMC6912964 DOI: 10.1161/jaha.119.013716] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Circulating proteins are exposed to vascular endothelial layer and influence their functions. Among them, adipsin is a member of the trypsin family of peptidases and is mainly secreted from adipocytes, monocytes, and macrophages, catalyzing the rate‐limiting step of the alternative complement pathway. However, its pathophysiological role in cardiovascular disease remains to be elucidated. Here, we examined whether serum adipsin levels have a prognostic impact in patients with coronary artery disease. Methods and Results In 370 consecutive patients undergoing diagnostic coronary angiography, we performed a cytokine array analysis for screening serum levels of 50 cytokines/chemokines and growth factors. Among them, classification and regression analysis identified adipsin as the best biomarker for prediction of their long‐term prognosis (median 71 months; interquartile range, 55–81 months). Kaplan–Meier curve showed that higher adipsin levels (≥400 ng/mL) were significantly associated with all‐cause death (hazard ratio [HR], 4.2; 95% CI, 1.7–10.6 [P<0.001]) and rehospitalization (HR, 2.4; 95% CI, 1.7–3.5 [P<0.001]). Interestingly, higher high‐sensitivity C‐reactive protein levels (≥1 mg/L) were significantly correlated with all‐cause death (HR, 3.2; 95% CI, 1.7–5.9 [P<0.001]) and rehospitalization (HR, 1.5, 95% CI, 1.1–1.9 [P<0.01]). Importantly, the combination of adipsin (≥400 ng/mL) and high‐sensitivity C‐reactive protein (≥1 mg/L) was more significantly associated with all‐cause death (HR, 21.0; 95% CI, 2.9–154.1 [P<0.001]). Finally, the receiver operating characteristic curve demonstrated that serum adipsin levels predict the death caused by acute myocardial infarction in patients with coronary artery disease (C‐statistic, 0.847). Conclusions These results indicate that adipsin is a novel biomarker that predicts all‐cause death and rehospitalization in patients with coronary artery disease, demonstrating the novel aspects of the alternative complementary system in the pathogenesis of coronary artery disease.
Collapse
Affiliation(s)
- Tomohiro Ohtsuki
- Department of Cardiovascular Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Kimio Satoh
- Department of Cardiovascular Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Toru Shimizu
- Department of Cardiovascular Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Shohei Ikeda
- Department of Cardiovascular Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Nobuhiro Kikuchi
- Department of Cardiovascular Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Taijyu Satoh
- Department of Cardiovascular Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Ryo Kurosawa
- Department of Cardiovascular Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Masamichi Nogi
- Department of Cardiovascular Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Shinichiro Sunamura
- Department of Cardiovascular Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Nobuhiro Yaoita
- Department of Cardiovascular Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Junichi Omura
- Department of Cardiovascular Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Tatsuo Aoki
- Department of Cardiovascular Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Shunsuke Tatebe
- Department of Cardiovascular Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Koichiro Sugimura
- Department of Cardiovascular Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Jun Takahashi
- Department of Cardiovascular Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Satoshi Miyata
- Department of Cardiovascular Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine Tohoku University Graduate School of Medicine Sendai Japan
| |
Collapse
|
105
|
Gumanova NG. Analytical complex of biochemical markers for preclinical diagnosis and prevention of cardiovascular diseases. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2019. [DOI: 10.15829/1728-8800-2019-5-117-127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Affiliation(s)
- N. G. Gumanova
- National Medical Research Center for Preventive Medicine
| |
Collapse
|
106
|
Xu M, Luo L, Du M, Tang L, Hu Y, Mei H. Fibrinogen levels are associated with bleeding in patients with primary immune thrombocytopenia. Platelets 2019; 31:763-770. [PMID: 31610686 DOI: 10.1080/09537104.2019.1678115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Min Xu
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lili Luo
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mengyi Du
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lu Tang
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu Hu
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Heng Mei
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
107
|
Wang PW, Lin TY, Hung YC, Chang WN, Yang PM, Chen MH, Yeh CT, Pan TL. Characterization of Fibrinogen as a Key Modulator in Patients with Wilson's Diseases with Functional Proteomic Tools. Int J Mol Sci 2019; 20:ijms20184528. [PMID: 31547461 PMCID: PMC6770682 DOI: 10.3390/ijms20184528] [Citation(s) in RCA: 3] [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: 08/12/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 12/26/2022] Open
Abstract
Wilson’s disease (WD) is an autosomal recessive disorder of copper metabolism caused by defects in the ATPase gene (ATP7B). The various clinical features result from the massive accumulation of copper in the liver, cornea and basal ganglia. Although WD can be effectively treated with proper medicine, this disease is difficult to clearly diagnose due to its indefinite symptoms. In the current study, we achieved a positive correlation between clinical symptoms and the enzymatic activity of ceruloplasmin in WD patients. Furthermore, proteome profiles of plasma as well as network analysis demonstrated that fibrinogen is a critical indicator which is significantly unregulated in WD subjects in comparison to healthy donors and closely linked to pathogenesis of WD. Here, we applied 2DE-immunoblots and immunohistochemistry to verify the protein level and localization in situ. The enhanced expression of fibrinogen in the plasma of WD subjects with respect to that of healthy controls and patients with distinct disorders was also confirmed by utilizing clinical samples. As expected, application of high dose of copper induced expression of fibrinogen, while knockdown of ceruloplasmin also resulted in upregulation of fibrinogen as well as elimination of superoxide dismutase (SOD), leading to increased oxidative stress in cells. In summary, the liver injury or oxidative stress induced by the progression of WD may account for the obvious increase of fibrinogen, which in turn triggers inflammatory responses and interferes coagulation cascades; this finding sheds light on the early detection and diagnosis of WD.
Collapse
Affiliation(s)
- Pei-Wen Wang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40447, Taiwan.
| | - Tung-Yi Lin
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan.
| | - Yu-Chiang Hung
- Department of Chinese Medicine, College of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung 83301, Taiwan.
| | - Wen-Neng Chang
- Departments of Neurology, College of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung 83301, Taiwan.
| | - Pei-Ming Yang
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11042, Taiwan.
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11042, Taiwan.
| | - Mu-Hong Chen
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- Department of Psychiatry, College of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 33375, Taiwan.
| | - Tai-Long Pan
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 33375, Taiwan.
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
- Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan 33302, Taiwan.
- Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan.
| |
Collapse
|
108
|
Relationship between fibrinogen levels and cardiovascular events in patients receiving percutaneous coronary intervention: a large single-center study. Chin Med J (Engl) 2019; 132:914-921. [PMID: 30958432 PMCID: PMC6595753 DOI: 10.1097/cm9.0000000000000181] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Background: It is currently unclear if fibrinogen is a risk factor for adverse events in patients receiving percutaneous coronary intervention (PCI) or merely serves as a marker of pre-existing comorbidities and other causal factors. We therefore investigated the association between fibrinogen levels and 2-year all-cause mortality, and compared the additional predictive value of adding fibrinogen to a basic model including traditional risk factors in patients receiving contemporary PCI. Methods: A total of 6293 patients undergoing PCI with measured baseline fibrinogen levels were enrolled from January to December 2013 in Fuwai Hospital. Patients were divided into three groups according to tertiles of baseline fibrinogen levels: low fibrinogen, <2.98 g/L; medium fibrinogen, 2.98 to 3.58 g/L; and high fibrinogen, ≥3.58 g/L. Independent predictors of 2-year clinical outcomes were determined by multivariate Cox proportional hazards regression modeling. The increased discriminative value of fibrinogen for predicting all-cause mortality was assessed using the C-statistic and integrated discrimination improvement (IDI). Results: The 2-year all-cause mortality rate was 1.2%. It was significantly higher in the high fibrinogen compared with the low and medium fibrinogen groups according to Kaplan-Meier analyses (1.7% vs. 0.9% and 1.7% vs. 1.0%, respectively; log-rank, P = 0.022). Fibrinogen was significantly associated with all-cause mortality according to multivariate Cox regression (hazard ratio 1.339, 95% confidence interval: 1.109–1.763, P = 0.005), together with traditional risk factors including age, sex, diabetes mellitus, left ventricular ejection fraction, creatinine clearance, and low-density lipoprotein cholesterol. The area under the curve for all-cause mortality in the basic model including traditional risk factors was 0.776, and this value increased to 0.787 when fibrinogen was added to the model (IDI = 0.003, Z = 0.140, P = 0.889). Conclusions: Fibrinogen is associated with 2-year all-cause mortality in patients receiving PCI, but provides no additional information over a model including traditional risk factors.
Collapse
|
109
|
Westberg H, Hedbrant A, Persson A, Bryngelsson IL, Johansson A, Ericsson A, Sjögren B, Stockfelt L, Särndahl E, Andersson L. Inflammatory and coagulatory markers and exposure to different size fractions of particle mass, number and surface area air concentrations in Swedish iron foundries, in particular respirable quartz. Int Arch Occup Environ Health 2019; 92:1087-1098. [PMID: 31165309 PMCID: PMC6814634 DOI: 10.1007/s00420-019-01446-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 05/28/2019] [Indexed: 01/06/2023]
Abstract
Purpose To study the relationship between inhalation of airborne particles and quartz in Swedish iron foundries and markers of inflammation and coagulation in blood. Methods Personal sampling of respirable dust and quartz was performed for 85 subjects in three Swedish iron foundries. Stationary measurements were used to study the concentrations of respirable dust and quartz, inhalable and total dust, PM10 and PM2.5, as well as the particle surface area and the particle number concentrations. Markers of inflammation, namely interleukins (IL-1β, IL-6, IL-8, IL-10 and IL-12), C-reactive protein, and serum amyloid A (SAA) were measured in plasma or serum, together with markers of coagulation including fibrinogen, factor VIII (FVIII), von Willebrand factor and d-dimer. Complete sampling was performed on the second or third day of a working week after a work-free weekend, and follow-up samples were collected 2 days later. A mixed model analysis was performed including sex, age, smoking, infections, blood group, sampling day and BMI as covariates. Results The average 8-h time-weighted average air concentrations of respirable dust and quartz were 0.85 mg/m3 and 0.052 mg/m3, respectively. Participants in high-exposure groups with respect to some of the measured particle types exhibited significantly elevated levels of SAA, fibrinogen and FVIII. Conclusions These observed relationships between particle exposure and inflammatory markers may indicate an increased risk of cardiovascular disease among foundry workers with high particulate exposure.
Collapse
Affiliation(s)
- Håkan Westberg
- Department of Occupational and Environmental Medicine, Faculty of Medicine and Health, Örebro University, 70182, Örebro, Sweden. .,Department of Medical Sciences, School of Medicine and Health, Örebro University, 701 82, Örebro, Sweden. .,Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, 701 82, Örebro, Sweden. .,Department of Occupational and Environmental Medicine, Örebro University Hospital, 701 85, Örebro, Sweden.
| | - Alexander Hedbrant
- Department of Medical Sciences, School of Medicine and Health, Örebro University, 701 82, Örebro, Sweden.,Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, 701 82, Örebro, Sweden
| | - Alexander Persson
- Department of Medical Sciences, School of Medicine and Health, Örebro University, 701 82, Örebro, Sweden.,Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, 701 82, Örebro, Sweden
| | - Ing-Liss Bryngelsson
- Department of Occupational and Environmental Medicine, Faculty of Medicine and Health, Örebro University, 70182, Örebro, Sweden
| | - Anders Johansson
- Department of Occupational and Environmental Medicine, Faculty of Medicine and Health, Örebro University, 70182, Örebro, Sweden
| | - Annette Ericsson
- Department of Occupational and Environmental Medicine, Faculty of Medicine and Health, Örebro University, 70182, Örebro, Sweden
| | - Bengt Sjögren
- Work Environment Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Leo Stockfelt
- Department of Occupational and Environmental Medicine, University of Gothenburg, PB 414, 405 30, Göteborg, Sweden
| | - Eva Särndahl
- Department of Medical Sciences, School of Medicine and Health, Örebro University, 701 82, Örebro, Sweden.,Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, 701 82, Örebro, Sweden
| | - Lena Andersson
- Department of Occupational and Environmental Medicine, Faculty of Medicine and Health, Örebro University, 70182, Örebro, Sweden.,Department of Medical Sciences, School of Medicine and Health, Örebro University, 701 82, Örebro, Sweden
| |
Collapse
|
110
|
Rinde LB, Morelli VM, Småbrekke B, Mathiesen EB, Løchen ML, Njølstad I, Wilsgaard T, Smith E, Rosendaal FR, Frazer KA, Braekkan SK, Hansen JB. Effect of prothrombotic genotypes on the risk of venous thromboembolism in patients with and without ischemic stroke. The Tromsø Study. J Thromb Haemost 2019; 17:749-758. [PMID: 30773804 DOI: 10.1111/jth.14410] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 02/09/2019] [Indexed: 11/27/2022]
Abstract
Essentials Prothrombotic genotypes may agument the risk of venous thromboembolism (VTE) after ischemic stroke. We studied this effect in a case-cohort study using a genetic risk score. In stroke patients, a one-category increase in the genetic risk score was associated with a 50% higher relative risk of VTE. The risk of VTE in stroke patients increased with an increasing number of risk alleles. SUMMARY: Background Patients with ischemic stroke have a transiently increased risk of subsequent venous thromboembolism (VTE). Prothrombotic genotypes may augment VTE risk under conditions of high thrombosis risk related to stroke. Aims To investigate the effect of prothrombotic genotypes in patients with ischemic stroke on the risk of VTE in a population-based case-cohort study. Methods Cases with incident VTE (n = 664) and a randomly selected age-weighted subcohort (n = 1817) were sampled from three surveys of the Tromsø Study (1994-2008). Participants were genotyped for ABO (rs8176719), F5 (rs6025), F2 (rs1799963), FGG (rs2066865) and F11 (rs2036914) single-nucleotide polymorphisms (SNPs). Cox regression models were used to calculate hazard ratios (HRs) for incident VTE according to individual SNPs and categories of risk alleles (5-SNP score; 0-1, 2, 3-4 and ≥ 5) in participants with and without ischemic stroke. Results There were 192 patients with incident stroke, of whom 43 developed VTE during a median of 15.2 years of follow-up. The risk alleles of individual SNPs augmented the elevated VTE risk brought about by ischemic stroke. In stroke patients, a one-category increase in the genetic risk score was associated with a 50% higher relative risk of overall VTE (HR 1.5, 95% confidence interval [CI] 1.3-1.8) and an 80% higher relative risk of provoked VTE (HR 1.8, 95% CI 1.5-2.1). Stroke patients with ≥ 5 risk alleles had a 12-fold (HR 11.7, 95% CI 4.1-33.3) higher relative risk of VTE than stroke-free participants with 0-1 risk alleles. Conclusions Prothrombotic genotypes increased the risk of VTE in stroke patients, and the risk increased with an increasing number of risk alleles.
Collapse
Affiliation(s)
- Ludvig B Rinde
- K. G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Vania M Morelli
- K. G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Birgit Småbrekke
- K. G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ellisiv B Mathiesen
- K. G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
- Brain and Circulation Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Maja-Lisa Løchen
- Epidemiology of Chronic Diseases Research Group, Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Inger Njølstad
- K. G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
- Epidemiology of Chronic Diseases Research Group, Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Tom Wilsgaard
- Epidemiology of Chronic Diseases Research Group, Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Erin Smith
- K. G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
- Department of Pediatrics and Rady's Children's Hospital, University of California, San Diego, La Jolla, CA, USA
| | - Frits R Rosendaal
- K. G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Kelly A Frazer
- K. G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
- Department of Pediatrics and Rady's Children's Hospital, University of California, San Diego, La Jolla, CA, USA
| | - Sigrid K Braekkan
- K. G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
- Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - John-Bjarne Hansen
- K. G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
- Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
111
|
Fontecha J, Calvo MV, Juarez M, Gil A, Martínez-Vizcaino V. Milk and Dairy Product Consumption and Cardiovascular Diseases: An Overview of Systematic Reviews and Meta-Analyses. Adv Nutr 2019; 10:S164-S189. [PMID: 31089735 PMCID: PMC6518146 DOI: 10.1093/advances/nmy099] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/01/2018] [Accepted: 10/26/2018] [Indexed: 12/20/2022] Open
Abstract
Milk and dairy products containing milk fat are major food sources of saturated fatty acids, which have been linked to increased risk of cardiovascular-related clinical outcomes such as cardiovascular disease (CVD), coronary heart disease (CHD), and stroke. Therefore, current recommendations by health authorities advise consumption of low-fat or fat-free milk. Today, these recommendations are seriously questioned by meta-analyses of both prospective cohort studies and randomized controlled trials (RCTs) reporting inconsistent results. The present study includes an overview of systematic reviews and meta-analyses of follow-up studies, an overview of meta-analyses involving RCTs, and an update on meta-analyses of RCTs (2013-2018) aiming to synthesize the evidence regarding the influence of dairy product consumption on the risk of major cardiovascular-related outcomes and how various doses of different dairy products affect the responses, as well as on selected biomarkers of cardiovascular disease risk, i.e., blood pressure and blood lipids. The search strategies for both designs were conducted in the MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials, Cochrane Database of Systematic Reviews, and Web of Science databases from their inception to April 2018. From the 31 full-text articles retrieved for cohort studies, 17 met the eligibility criteria. The pooled risk ratio estimated for the association between the consumption of different dairy products at different dose-responses and cardiovascular outcomes (CVD, CHD, and stroke) showed a statistically significant negative association with RR values <1, or did not find evidence of significant association. The overview of 12 meta-analyses involving RCTs as well as the updated meta-analyses of RCTs did not result in significant changes on risk biomarkers such as systolic and diastolic blood pressure and total cholesterol and LDL cholesterol. Therefore, the present study states that the consumption of total dairy products, with either regular or low fat content, does not adversely affect the risk of CVD.
Collapse
Affiliation(s)
- Javier Fontecha
- Department of Bioactivity and Food Analysis, Food Lipid Biomarkers and Health Group, Institute of Food Science Research, CIAL (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain,Address correspondence to JF (e-mail: )
| | - Maria Visitación Calvo
- Department of Bioactivity and Food Analysis, Food Lipid Biomarkers and Health Group, Institute of Food Science Research, CIAL (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuela Juarez
- Department of Bioactivity and Food Analysis, Food Lipid Biomarkers and Health Group, Institute of Food Science Research, CIAL (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Angel Gil
- Department of Biochemistry and Molecular Biology II, School of Pharmacy,Institute of Nutrition and Food Technology “José Mataix,” Biomedical Research Center, University of Granada, Granada, Spain,Instituto de Investigación Biosanitaria ibs GRANADA, Complejo Hospitalario Universitario de Granada, Granada, Spain,CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, Madrid, Spain
| | - Vicente Martínez-Vizcaino
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain,Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| |
Collapse
|
112
|
Soto-Méndez MJ, Rangel-Huerta OD, Ruiz-López MD, Martínez de Victoria E, Anguita-Ruiz A, Gil A. Role of Functional Fortified Dairy Products in Cardiometabolic Health: A Systematic Review and Meta-analyses of Randomized Clinical Trials. Adv Nutr 2019; 10:S251-S271. [PMID: 31089744 PMCID: PMC6518140 DOI: 10.1093/advances/nmz001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/26/2018] [Indexed: 12/11/2022] Open
Abstract
There is insufficient evidence on the role of functional fortified dairy products in improving health and in preventing risk factors associated with noncommunicable chronic diseases. This systematic review was conducted to summarize effects of the consumption of fortified dairy products on biomarkers of cardiometabolic risk. MEDLINE and SCOPUS databases were used to perform searches to include studies published up to 30 April 2018. Randomized clinical trials with human subjects consuming dairy products fortified with phytosterols, FAs, vitamins or minerals and relating this consumption with cardiometabolic health were included in this review. Risk of bias assessment according to Cochrane guidelines was performed to determine the quality of the trials. Forty-one studies were finally selected for this synthesis; the selected studies tested dairy products fortified with the following nutrients and bioactive components: phytosterols (n = 31), FAs (n = 8), and vitamin D (n = 2). We found that the consumption of phytosterol-fortified dairy, led to an overall LDL cholesterol reduction of -0.36 (-0.41, -0.31) mmol/L, P < 0.001; this decrease was mainly related to the dosage. Likewise, consumption of ω-3 FA-fortified dairy products resulted in a plasma LDL cholesterol reduction of -0.18 (-0.27, -0.09) mmol/L as well as a decrease of -0.18 (-0.32, -0.05) mmol/L in triacylglycerols (TG). Performing meta-analyses of the consumption of dairy products fortified with vitamin D or FAs other than ω-3 FAs and biomarkers of cardiometabolic risk was not possible because of the few available publications. Our results indicate that consumption of dairy products fortified with phytosterols and ω-3 FAs can lead to a reduction of LDL cholesterol and consumption of fortified dairy products fortified with ω-3 FAs can reduce TG concentration. However, more studies with homogeneous designs are needed to determine the advantages of using dairy products as fortification vehicles to prevent cardiometabolic risk.
Collapse
Affiliation(s)
| | - Oscar D Rangel-Huerta
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - María D Ruiz-López
- Iberoamerican Nutrition Foundation –FINUT–, Granada, Spain
- Department of Nutrition and Food Sciences, School of Pharmacy
- Institute of Nutrition and Food Technology “José Mataix,” Biomedical Research Center, University of Granada, Granada, Spain
| | - Emilio Martínez de Victoria
- Iberoamerican Nutrition Foundation –FINUT–, Granada, Spain
- Department of Physiology, School of Sciences
- Institute of Nutrition and Food Technology “José Mataix,” Biomedical Research Center, University of Granada, Granada, Spain
| | - Augusto Anguita-Ruiz
- Institute of Nutrition and Food Technology “José Mataix,” Biomedical Research Center, University of Granada, Granada, Spain
- Biomedical Research Institute ibs GRANADA, University Hospital Complex in Granada, Granada, Spain
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, Madrid, Spain
| | - Angel Gil
- Iberoamerican Nutrition Foundation –FINUT–, Granada, Spain
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix,” Biomedical Research Center, University of Granada, Granada, Spain
- Biomedical Research Institute ibs GRANADA, University Hospital Complex in Granada, Granada, Spain
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
113
|
Zhang L, Xu C, Liu J, Bai X, Li R, Wang L, Zhou J, Wu Y, Yuan Z. Baseline plasma fibrinogen is associated with haemoglobin A1c and 2-year major adverse cardiovascular events following percutaneous coronary intervention in patients with acute coronary syndrome: a single-centre, prospective cohort study. Cardiovasc Diabetol 2019; 18:52. [PMID: 31014348 PMCID: PMC6480802 DOI: 10.1186/s12933-019-0858-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/10/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Despite revascularisation, a large proportion of acute coronary syndrome (ACS) patients continue to experience major adverse cardiovascular events (MACEs), which are worsened by diabetes mellitus (DM). Fibrinogen (FIB) is a risk factor for MACEs in coronary artery disease and often elevated in DM. However, the relationships between FIB, glucose metabolism (haemoglobin A1c [HbA1c] and fasting blood glucose [FBG]) and MACEs following percutaneous coronary intervention (PCI) in DM, non-DM or whole patients with ACS remains unknown. METHODS A total of 411 ACS patients undergoing PCI were enrolled in this study. We compared baseline FIB levels between DM (n = 103) and non-DM (n = 308) patients and divided participants into three groups according to FIB level, i.e. FIB-L, FIB-M and FIB-H, to compare baseline characteristics and MACEs. Linear regression analysis of the relationship between glucose metabolism and FIB, Cox regression, survival and landmark analyses of MACEs were also performed over a median of 27.55 months of follow-up. RESULTS Patients with DM had higher FIB levels than non-DM patients (3.56 ± 0.99 mg/dL vs. 3.34 ± 0.80 mg/dL, P < 0.05). HbA1c and FBG were significantly positively correlated with FIB in whole and DM patients but not in non-DM patients (all P < 0.05). Compared with the FIB-L group, the FIB-M (hazard ratio [HR] 1.797, 95% CI 1.117-2.892, P = 0.016) and FIB-H (HR 1.664, 95% CI 1.002-2.763, P = 0.049) groups were associated with higher MACEs in whole; the FIB-M (HR 7.783, 95% CI 1.012-59.854, P = 0.049) was associated with higher MACEs in DM patients. FIB was not associated with MACEs in non-DM patients. During landmark analysis, FIB showed better predictive value for MACEs after PCI in the first 30 months of follow up than in the subsequent period. CONCLUSION In this study from China, FIB was positively associated with glucose metabolism (HbA1c and FBG) in whole and DM populations with ACS. Moreover, elevated baseline FIB levels may be an important and independent predictor of MACEs following PCI, especially amongst those with DM. However, as the follow-up period increased, the baseline FIB levels lost their ability to predict MACEs.
Collapse
Affiliation(s)
- Lisha Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Rd., Xi'an, 710061, Shaanxi, People's Republic of China
| | - Chenbo Xu
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Rd., Xi'an, 710061, Shaanxi, People's Republic of China
| | - Junhui Liu
- Department of Clinical Laboratory, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Xiaofang Bai
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Rd., Xi'an, 710061, Shaanxi, People's Republic of China
| | - Ruifeng Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Rd., Xi'an, 710061, Shaanxi, People's Republic of China
| | - Lijun Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Rd., Xi'an, 710061, Shaanxi, People's Republic of China
| | - Juan Zhou
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Rd., Xi'an, 710061, Shaanxi, People's Republic of China.,Key Laboratory of Molecular Cardiology, Shaanxi Province, Xi'an, Shaanxi, People's Republic of China
| | - Yue Wu
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Rd., Xi'an, 710061, Shaanxi, People's Republic of China. .,Key Laboratory of Molecular Cardiology, Shaanxi Province, Xi'an, Shaanxi, People's Republic of China.
| | - Zuyi Yuan
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Rd., Xi'an, 710061, Shaanxi, People's Republic of China. .,Key Laboratory of Molecular Cardiology, Shaanxi Province, Xi'an, Shaanxi, People's Republic of China. .,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, People's Republic of China.
| |
Collapse
|
114
|
Wang J, Jia L, Li X, Jin S, Li X, Liu F, Shan C, Zhang Y, Yang Y. New Insights into the Association between Fibrinogen and Coronary Atherosclerotic Plaque Vulnerability: An Intravascular Optical Coherence Tomography Study. Cardiovasc Ther 2019; 2019:8563717. [PMID: 31772619 PMCID: PMC6740041 DOI: 10.1155/2019/8563717] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 01/31/2019] [Accepted: 02/24/2019] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Fibrinogen levels have been associated with coronary plaque vulnerability in experimental studies. However, it has yet to be determined if serum fibrinogen levels are independently associated with coronary plaque vulnerability as detected by optical coherence tomography (OCT) in patients with coronary heart disease. METHODS Patients with coronary heart disease (CHD) who underwent coronary angiography and OCT in our department from January 2015 to August 2018 were included in this study. Coronary lesions were categorized as ruptured plaque, nonruptured with thin-cap fibroatheroma (TCFA), and nonruptured and non-TCFA. Presence of ruptured plaque and nonruptured with TCFA was considered to be vulnerable lesions. Determinants of coronary vulnerability were evaluated by multivariable logistic regression analyses. RESULTS A total of 154 patients were included in this study; 17 patients had ruptured plaques, 15 had nonruptured plaques with TCFA, and 122 had nonruptured plaques with non-TCFA. Results of univariate analyses showed that being male, diabetes, current smoking, high body mass index (BMI), and clinical diagnosis of acute coronary syndrome (ACS) were associated with coronary vulnerability. No significant differences were detected in patient characteristics, coronary angiographic findings, and OCT results between patients with higher and normal fibrinogen. Results of multivariate logistic analyses showed that diabetes and ACS were associated with TCFA, while diabetes, higher BMI, and ACS were associated with plaque rupture. CONCLUSIONS Diabetes, higher BMI, and ACS are independently associated with coronary vulnerability as detected by OCT. Serum fibrinogen was not associated with coronary vulnerability in our cohort.
Collapse
Affiliation(s)
- Jun Wang
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Lu Jia
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Xing Li
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Siyu Jin
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Xiaomei Li
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Fen Liu
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Chunfang Shan
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Yu Zhang
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Yining Yang
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| |
Collapse
|
115
|
Causes of Death in Implant Patients Treated in the Edentulous Jaw: A Comparison between 2098 Deceased Patients and the Swedish National Cause of Death Register. Int J Dent 2019; 2019:7315081. [PMID: 30984265 PMCID: PMC6432731 DOI: 10.1155/2019/7315081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/14/2019] [Indexed: 01/11/2023] Open
Abstract
Background Previous research has reported an association between tooth loss and patient mortality, while the cause of death has not been elucidated. Objective The purpose was to describe and compare the cause of death in implant patients treated consecutively in the edentulous arch with a reference population. Methods Altogether, 3902 patients were included between 1986 and 2014. Data on the causes of death for deceased patients were compared to the Swedish National Cause of Death Register for a comparable time period. Standardised mortality ratios (SMRs) were calculated based on gender and age and tested for statistical significance. Results Most deceased patients (2,098) died from diseases in the circulatory system (CVD; 42%) and from cancers (26%). SMR indicated a generally increased mortality (total group) compared to the reference population during inclusion (P < 0.05; 1986–2014). Patients treated early (1986–1996) showed a lower SMR compared to patients treated later (P < 0.05; 1997–2014) especially related to CVDs. Younger patients (<60 years at surgery) showed an increased mortality due to CVDs when treated late (1997–2014; SMR = 5.4, P < 0.05). Elderly patients (>79 years at surgery) showed a significantly lower mortality in almost all observed causes of death (1986–2014; P < 0.05) with also a significantly lower mortality due to CVDs during the early period (1986–1996; SMR = 0.3, P < 0.05). Conclusion An overall increased mortality was observed for the edentulous implant patient compared to the reference population. Elderly patients (>79 years) showed significantly lower mortality for all causes of death independent of the time period of implant surgery. Younger patients (<60 years) present an increased risk for early mortality related to CVD. SMR for all causes of death increased for patients treated late (1997–2014) as compared to patients treated early (1986–1996).
Collapse
|
116
|
Loxham M, Nieuwenhuijsen MJ. Health effects of particulate matter air pollution in underground railway systems - a critical review of the evidence. Part Fibre Toxicol 2019; 16:12. [PMID: 30841934 PMCID: PMC6404319 DOI: 10.1186/s12989-019-0296-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 02/21/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Exposure to ambient airborne particulate matter is a major risk factor for mortality and morbidity, associated with asthma, lung cancer, heart disease, myocardial infarction, and stroke, and more recently type 2 diabetes, dementia and loss of cognitive function. Less is understood about differential effects of particulate matter from different sources. Underground railways are used by millions of people on a daily basis in many cities. Poor air exchange with the outside environment means that underground railways often have an unusually high concentration of airborne particulate matter, while a high degree of railway-associated mechanical activity produces particulate matter which is physicochemically highly distinct from ambient particulate matter. The implications of this for the health of exposed commuters and employees is unclear. MAIN BODY A literature search found 27 publications directly assessing the potential health effects of underground particulate matter, including in vivo exposure studies, in vitro toxicology studies, and studies of particulate matter which might be similar to that found in underground railways. The methodology, findings, and conclusions of these studies were reviewed in depth, along with further publications directly relevant to the initial search results. In vitro studies suggest that underground particulate matter may be more toxic than exposure to ambient/urban particulate matter, especially in terms of endpoints related to reactive oxygen species generation and oxidative stress. This appears to be predominantly a result of the metal-rich nature of underground particulate matter, which is suggestive of increased health risks. However, while there are measureable effects on a variety of endpoints following exposure in vivo, there is a lack of evidence for these effects being clinically significant as may be implied by the in vitro evidence. CONCLUSION There is little direct evidence that underground railway particulate matter exposure is more harmful than ambient particulate matter exposure. This may be due to disparities between in vivo exposures and in vitro models, and differences in exposure doses, as well as statistical under powering of in vivo studies of chronic exposure. Future research should focus on outcomes of chronic in vivo exposure, as well as further work to understand mechanisms and potential biomarkers of exposure.
Collapse
Affiliation(s)
- Matthew Loxham
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Mailpoint 888, Level F, University Hospital Southampton, Tremona Road, Southampton, SO16 6YD, UK. .,NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK. .,Institute for Life Sciences, University of Southampton, Southampton, UK. .,Southampton Marine and Maritime Institute, University of Southampton, Southampton, UK.
| | - Mark J Nieuwenhuijsen
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| |
Collapse
|
117
|
Li T, Wang F, Peng R, Pei S, Hou Z, Lu B, Cong X, Chen X. Sex-related differences in the association between plasma fibrinogen and non-calcified or mixed coronary atherosclerotic plaques. Biol Sex Differ 2018; 9:51. [PMID: 30518417 PMCID: PMC6282270 DOI: 10.1186/s13293-018-0210-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 11/22/2018] [Indexed: 12/19/2022] Open
Abstract
Background Plasma fibrinogen (FIB) has been demonstrated to be a risk factor for cardiovascular disease. Patients with non-calcified plaque (NCP) or mix plaque (MP) have a higher risk of poor outcomes. However, the association between FIB and the presence of NCP or MP (NCP/MP) remains unclear, and if present, whether sex has any impact on this association remains unknown. The aim of this study was to investigate the role of FIB in predicting the presence of NCP/MP and evaluate whether sex has any impact on this association. Methods A total of 329 subjects were recruited, and the clinical and laboratory data were collected. Plasma FIB was detected by enzyme-linked immunosorbent assay. According to whether they had coronary atherosclerotic plaques and the characteristics of the most stenotic plaque, we divided them into three groups: no plaque (NP), calcified plaque (CP), and NCP/MP. Results Patients with NCP/MP had significantly higher FIB level in females, but not in males. Multiple logistic regression analysis showed that FIB was an independent risk factor for the presence of NCP/MP (odds ratio [OR] = 3.677, 95% CI 1.539–8.785, P = 0.003) in females. Receiver operating characteristic (ROC) curve analysis showed that the optimal cut-off value FIB for predicting the presence of NCP/MP was 3.41 g/L (area under curve [AUC] = 0.73, 95% CI 0.63–0.82, P < 0.001) in females. Conclusions FIB is independently associated with the presence of NCP/MP in females, but not in males. These results suggest that the potential significance of FIB-lowering regimens in females with NCP/MP.
Collapse
Affiliation(s)
- Tiewei Li
- Department of Clinical Laboratory Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishi Street, Xi-Cheng District, Beijing, 100037, China
| | - Fang Wang
- Department of Clinical Laboratory Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishi Street, Xi-Cheng District, Beijing, 100037, China.
| | - Rui Peng
- Department of Clinical Laboratory Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishi Street, Xi-Cheng District, Beijing, 100037, China
| | - Shengqiang Pei
- Department of Clinical Laboratory Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishi Street, Xi-Cheng District, Beijing, 100037, China
| | - Zhihui Hou
- Department of Radiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Bin Lu
- Department of Radiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiangfeng Cong
- Department of Clinical Laboratory Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishi Street, Xi-Cheng District, Beijing, 100037, China
| | - Xi Chen
- Department of Clinical Laboratory Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishi Street, Xi-Cheng District, Beijing, 100037, China.
| |
Collapse
|
118
|
Nakabayashi K, Jujo K, Furuki Y, Ishida I, Ando H, Shimizu M, Hagiwara N, Saito K. Variations in the eicosapentaenoic acid-arachidonic acid ratio associated with age in acute myocardial infarction patients undergoing primary percutaneous coronary intervention. Heart Vessels 2018; 34:755-762. [PMID: 30430295 DOI: 10.1007/s00380-018-1302-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 11/09/2018] [Indexed: 12/17/2022]
Abstract
Acute myocardial infarction (AMI) is a life-threatening disease, and its incidence has been increasing even in the young population. Although a low eicosapentaenoic acid (EPA)-arachidonic acid (AA) ratio is associated with an increased risk of coronary artery disease, the effect of age on EPA/AA ratios in AMI patients remains unclear. This study aimed to clarify the independent polyunsaturated fatty acid (PUFA)-related determinants of age in younger and older AMI patients. A total of 153 consecutive patients who underwent primary percutaneous coronary interventions (PCIs) for de novo AMIs were enrolled in this study. Patients' background data, including PUFA and lipid profiles during PCI, were evaluated retrospectively. The EPA/AA ratio correlated positively with age (r = 0.21; P = 0.011) and increased markedly from age 60 years. Patients aged < 60 years (n = 35) had a lower mean EPA/AA ratio (0.25 ± 0.16) than patients aged ≥ 60 years (n = 118) (0.38 ± 0.25) (P < 0.001). The AA level was more dependent on age than on EPA level (r = - 0.34, P < 0.001 vs. r = 0.12, P = 0.16). The multivariate analysis revealed that a 0.1 EPA/AA ratio increase (odds ratio 1.50; 95% confidence interval 1.09-2.06), body mass index, triglyceride level, and aspirin administration were independently associated with the age stratification of AMI patients. The EPA/AA ratio was higher in younger AMI patients who have undergone primary PCIs than in older patients. Younger population at risk for AMI should be managed with multiple interventions including PUFA profiling.
Collapse
Affiliation(s)
- Keisuke Nakabayashi
- Department of Cardiology, Kasukabe Chuo General Hospital, Saitama, Japan.,Department of Cardiology, Nishiarai Heart Center Hospital, Tokyo, Japan.,Department of Cardiology, Tokyo Women's Medical University, 8-1 Kawadacho, Shinjuku-ku, Tokyo, 162-0054, Japan
| | - Kentaro Jujo
- Department of Cardiology, Nishiarai Heart Center Hospital, Tokyo, Japan. .,Department of Cardiology, Tokyo Women's Medical University, 8-1 Kawadacho, Shinjuku-ku, Tokyo, 162-0054, Japan.
| | - Yuho Furuki
- Department of Cardiology, Nishiarai Heart Center Hospital, Tokyo, Japan
| | - Issei Ishida
- Department of Cardiology, Tokyo Women's Medical University, 8-1 Kawadacho, Shinjuku-ku, Tokyo, 162-0054, Japan
| | - Hiroshi Ando
- Department of Cardiology, Kasukabe Chuo General Hospital, Saitama, Japan
| | - Minoru Shimizu
- Department of Cardiology, Kasukabe Chuo General Hospital, Saitama, Japan
| | - Nobuhisa Hagiwara
- Department of Cardiology, Tokyo Women's Medical University, 8-1 Kawadacho, Shinjuku-ku, Tokyo, 162-0054, Japan
| | - Katsumi Saito
- Department of Cardiology, Nishiarai Heart Center Hospital, Tokyo, Japan
| |
Collapse
|
119
|
Makkar H, Reynolds MA, Wadhawan A, Dagdag A, Merchant AT, Postolache TT. Periodontal, metabolic, and cardiovascular disease: Exploring the role of inflammation and mental health. Pteridines 2018; 29:124-163. [PMID: 30705520 PMCID: PMC6350811 DOI: 10.1515/pteridines-2018-0013] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Previous evidence connects periodontal disease, a modifiable condition affecting a majority of Americans, with metabolic and cardiovascular morbidity and mortality. This review focuses on the likely mediation of these associations by immune activation and their potential interactions with mental illness. Future longitudinal, and ideally interventional studies, should focus on reciprocal interactions and cascading effects, as well as points for effective preventative and therapeutic interventions across diagnostic domains to reduce morbidity, mortality and improve quality of life.
Collapse
Affiliation(s)
- Hina Makkar
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mark A Reynolds
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Abhishek Wadhawan
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Aline Dagdag
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Anwar T Merchant
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Teodor T Postolache
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Denver, CO 80220, USA; Mental Illness Research, Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 5, VA Capitol Health Care Network, Baltimore, MD 21201, USA,
| |
Collapse
|
120
|
Alman AC, Talton JW, Wadwa RP, Urbina EM, Dolan LM, Hamman RF, D'Agostino RB, Marcovina SM, Dabelea DM. Inflammation, adiposity, and progression of arterial stiffness in adolescents with type 1 diabetes: The SEARCH CVD Study. J Diabetes Complications 2018; 32:995-999. [PMID: 30209019 PMCID: PMC6174105 DOI: 10.1016/j.jdiacomp.2018.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 12/28/2017] [Accepted: 08/06/2018] [Indexed: 11/30/2022]
Abstract
AIMS We examined the association between inflammation and progression of arterial stiffness in a population of youth with type 1 diabetes (T1D). METHODS A total of 287 youth with T1D (median age 13 years) from SEARCH CVD, an ancillary study to the SEARCH for Diabetes in Youth, were included. Markers of inflammation (CRP, IL-6, fibrinogen, leptin, and adiponectin) and measures of pulse wave velocity (PWV) of the arm (PWV-R), trunk (PWV-T), and lower extremity (PWV-LE) were measured at baseline. Measures of PWV were repeated approximately five years later. RESULTS PWV-R (0.50 m/s), PWV-T (0.65 m/s), and PWV-LE (1.0 m/s) significantly increased over the follow-up (p < 0.001 for each). A significant interaction was found between waist circumference and fibrinogen (p = 0.036) on the progression of PWV-T, suggesting that fibrinogen is more strongly associated with PWV progression in lean participants. CONCLUSIONS Improved understanding of adiposity, inflammation, and functional changes in the vascular system in patients with T1D is crucial.
Collapse
Affiliation(s)
- Amy C Alman
- Department of Epidemiology and Biostatistics, College of Public Health, University of South Florida, USA.
| | - Jennifer W Talton
- Department of Biostatistical Sciences, Wake Forest School of Medicine, USA
| | - R Paul Wadwa
- Barbara Davis Center, University of Colorado Denver, USA
| | - Elaine M Urbina
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, USA
| | - Lawrence M Dolan
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, USA
| | - Richard F Hamman
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, USA
| | - Ralph B D'Agostino
- Department of Biostatistical Sciences, Wake Forest School of Medicine, USA
| | - Santica M Marcovina
- Department of Metabolism, Endocrinology and Nutrition, University of Washington, USA
| | - Dana M Dabelea
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, USA
| |
Collapse
|
121
|
Ward-Caviness CK, Huffman JE, Everett K, Germain M, van Dongen J, Hill WD, Jhun MA, Brody JA, Ghanbari M, Du L, Roetker NS, de Vries PS, Waldenberger M, Gieger C, Wolf P, Prokisch H, Koenig W, O'Donnell CJ, Levy D, Liu C, Truong V, Wells PS, Trégouët DA, Tang W, Morrison AC, Boerwinkle E, Wiggins KL, McKnight B, Guo X, Psaty BM, Sotoodenia N, Boomsma DI, Willemsen G, Ligthart L, Deary IJ, Zhao W, Ware EB, Kardia SLR, Van Meurs JBJ, Uitterlinden AG, Franco OH, Eriksson P, Franco-Cereceda A, Pankow JS, Johnson AD, Gagnon F, Morange PE, de Geus EJC, Starr JM, Smith JA, Dehghan A, Björck HM, Smith NL, Peters A. DNA methylation age is associated with an altered hemostatic profile in a multiethnic meta-analysis. Blood 2018; 132:1842-1850. [PMID: 30042098 PMCID: PMC6202911 DOI: 10.1182/blood-2018-02-831347] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 07/01/2018] [Indexed: 01/25/2023] Open
Abstract
Many hemostatic factors are associated with age and age-related diseases; however, much remains unknown about the biological mechanisms linking aging and hemostatic factors. DNA methylation is a novel means by which to assess epigenetic aging, which is a measure of age and the aging processes as determined by altered epigenetic states. We used a meta-analysis approach to examine the association between measures of epigenetic aging and hemostatic factors, as well as a clotting time measure. For fibrinogen, we performed European and African ancestry-specific meta-analyses which were then combined via a random effects meta-analysis. For all other measures we could not estimate ancestry-specific effects and used a single fixed effects meta-analysis. We found that 1-year higher extrinsic epigenetic age as compared with chronological age was associated with higher fibrinogen (0.004 g/L/y; 95% confidence interval, 0.001-0.007; P = .01) and plasminogen activator inhibitor 1 (PAI-1; 0.13 U/mL/y; 95% confidence interval, 0.07-0.20; P = 6.6 × 10-5) concentrations, as well as lower activated partial thromboplastin time, a measure of clotting time. We replicated PAI-1 associations using an independent cohort. To further elucidate potential functional mechanisms, we associated epigenetic aging with expression levels of the PAI-1 protein encoding gene (SERPINE1) and the 3 fibrinogen subunit-encoding genes (FGA, FGG, and FGB) in both peripheral blood and aorta intima-media samples. We observed associations between accelerated epigenetic aging and transcription of FGG in both tissues. Collectively, our results indicate that accelerated epigenetic aging is associated with a procoagulation hemostatic profile, and that epigenetic aging may regulate hemostasis in part via gene transcription.
Collapse
Affiliation(s)
- Cavin K Ward-Caviness
- Institute of Epidemiology II, Helmholtz Center of Munich, Neuherberg, Germany
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Chapel Hill, NC
| | - Jennifer E Huffman
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA
- The Framingham Heart Study, Framingham, MA
- Center for Population Genomics, Boston VA Healthcare System, Jamaica Plain, MA
| | - Karl Everett
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Marine Germain
- Sorbonne Universités, UPMC University Paris 06, INSERM UMR_S 1166, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Jenny van Dongen
- Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - W David Hill
- Centre for Cognitive Ageing and Cognitive Epidemiology and
- Department of Psychology, University of Edinburgh, Edinburgh, United Kingdom
| | - Min A Jhun
- Department of Epidemiology, University of Michigan, Ann Arbor, MI
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Lei Du
- Cardiovascular Medicine Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Nicholas S Roetker
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Melanie Waldenberger
- Institute of Epidemiology II, Helmholtz Center of Munich, Neuherberg, Germany
- Research Unit of Molecular Epidemiology and
| | | | - Petra Wolf
- Institue of Human Genetics, Helmholtz Center of Munich, Neuherberg, Germany
| | - Holger Prokisch
- Institue of Human Genetics, Helmholtz Center of Munich, Neuherberg, Germany
- Institute fur Humangenetik, Technische Univeritat Munchen, Munich, Germany
| | - Wolfgang Koenig
- Department of Internal Medicine II-Cardiology, University of Ulm Medical Center, Ulm, Germany
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- German Centre for Cardiovascular Research, Munich Heart Alliance, Munich, Germany
| | - Christopher J O'Donnell
- The Framingham Heart Study, Framingham, MA
- Cardiology Section Administration, Boston VA Healthcare System, West Roxbury, MA
| | - Daniel Levy
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA
- The Framingham Heart Study, Framingham, MA
| | - Chunyu Liu
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA
- The Framingham Heart Study, Framingham, MA
| | - Vinh Truong
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Philip S Wells
- Department of Medicine, University of Ottawa and Ottawa Hospital Research Institute, Ottawa, Canada
| | - David-Alexandre Trégouët
- Sorbonne Universités, UPMC University Paris 06, INSERM UMR_S 1166, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Weihong Tang
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Barbara McKnight
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Xiuqing Guo
- Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrence, CA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
- Department of Epidemiology and
- Department of Health Services, University of Washington, Seattle, WA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA
| | - Nona Sotoodenia
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
- Division of Cardiology, University of Washington, Seattle, WA
| | - Dorret I Boomsma
- Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Gonneke Willemsen
- Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Lannie Ligthart
- Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology and
- Department of Psychology, University of Edinburgh, Edinburgh, United Kingdom
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI
| | - Erin B Ware
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI
| | | | - Joyce B J Van Meurs
- Department of Internal Medicine, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Andre G Uitterlinden
- Department of Internal Medicine, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Oscar H Franco
- Department of Epidemiology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Per Eriksson
- Cardiovascular Medicine Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Anders Franco-Cereceda
- Cardiothoracic Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - James S Pankow
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Andrew D Johnson
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA
- The Framingham Heart Study, Framingham, MA
| | - France Gagnon
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Pierre-Emmanuel Morange
- Laboratory of Hematology, La Timone Hospital, Marseille, France
- INSERM UMR_S 1062, Nutrition Obesity and Risk of Thrombosis, Center for CardioVascular and Nutrition Research, Aix-Marseille University, Marseille, France
| | - Eco J C de Geus
- Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health, VU Medical Center, Amsterdam, The Netherlands
| | - John M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology and
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, United Kingdom; and
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, MI
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Hanna M Björck
- Cardiovascular Medicine Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Nicholas L Smith
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Epidemiology and
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA
- Seattle Epidemiologic Research and Information Center, Office of Research and Development, Department of Veterans Affairs, Seattle, WA
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Center of Munich, Neuherberg, Germany
| |
Collapse
|
122
|
Xu XR, Wang Y, Adili R, Ju L, Spring CM, Jin JW, Yang H, Neves MAD, Chen P, Yang Y, Lei X, Chen Y, Gallant RC, Xu M, Zhang H, Song J, Ke P, Zhang D, Carrim N, Yu SY, Zhu G, She YM, Cyr T, Fu W, Liu G, Connelly PW, Rand ML, Adeli K, Freedman J, Lee JE, Tso P, Marchese P, Davidson WS, Jackson SP, Zhu C, Ruggeri ZM, Ni H. Apolipoprotein A-IV binds αIIbβ3 integrin and inhibits thrombosis. Nat Commun 2018; 9:3608. [PMID: 30190457 PMCID: PMC6127106 DOI: 10.1038/s41467-018-05806-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 07/19/2018] [Indexed: 12/29/2022] Open
Abstract
Platelet αIIbβ3 integrin and its ligands are essential for thrombosis and hemostasis, and play key roles in myocardial infarction and stroke. Here we show that apolipoprotein A-IV (apoA-IV) can be isolated from human blood plasma using platelet β3 integrin-coated beads. Binding of apoA-IV to platelets requires activation of αIIbβ3 integrin, and the direct apoA-IV-αIIbβ3 interaction can be detected using a single-molecule Biomembrane Force Probe. We identify that aspartic acids 5 and 13 at the N-terminus of apoA-IV are required for binding to αIIbβ3 integrin, which is additionally modulated by apoA-IV C-terminus via intra-molecular interactions. ApoA-IV inhibits platelet aggregation and postprandial platelet hyperactivity. Human apoA-IV plasma levels show a circadian rhythm that negatively correlates with platelet aggregation and cardiovascular events. Thus, we identify apoA-IV as a novel ligand of αIIbβ3 integrin and an endogenous inhibitor of thrombosis, establishing a link between lipoprotein metabolism and cardiovascular diseases. Activation of integrin αIIbβ3 at the surface of platelets is required for their aggregation and for thrombus formation. Here Xu et al. identify apolipoprotein A-IV as a novel ligand for platelet αIIbβ3 integrin, and find it inhibits platelet aggregation and thrombosis.
Collapse
Affiliation(s)
- Xiaohong Ruby Xu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada, M5S 1A1.,Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8.,Department of Acupuncture and Moxibustion, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, P.R. China, 510120.,Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P.R. China, 510000
| | - Yiming Wang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada, M5S 1A1.,Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8.,Canadian Blood Services Centre for Innovation, Toronto, ON, Canada, M5G 2M1
| | - Reheman Adili
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8
| | - Lining Ju
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA, 30332.,Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA, 30332.,Heart Research Institute, and Charles Perkins Centre, The University of Sydney, Camperdown, Australia, 2006
| | - Christopher M Spring
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8
| | - Joseph Wuxun Jin
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8.,Canadian Blood Services Centre for Innovation, Toronto, ON, Canada, M5G 2M1
| | - Hong Yang
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8.,Canadian Blood Services Centre for Innovation, Toronto, ON, Canada, M5G 2M1
| | - Miguel A D Neves
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8
| | - Pingguo Chen
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8.,Canadian Blood Services Centre for Innovation, Toronto, ON, Canada, M5G 2M1
| | - Yan Yang
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8.,Canadian Blood Services Centre for Innovation, Toronto, ON, Canada, M5G 2M1
| | - Xi Lei
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8
| | - Yunfeng Chen
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA, 30332.,Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA, 30332
| | - Reid C Gallant
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada, M5S 1A1.,Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8
| | - Miao Xu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada, M5S 1A1.,Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8
| | - Hailong Zhang
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8
| | - Jina Song
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8.,Canadian Blood Services Centre for Innovation, Toronto, ON, Canada, M5G 2M1
| | - Peifeng Ke
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P.R. China, 510000.,Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, P.R. China, 510120
| | - Dan Zhang
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8.,Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P.R. China, 510000
| | - Naadiya Carrim
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8.,Canadian Blood Services Centre for Innovation, Toronto, ON, Canada, M5G 2M1
| | - Si-Yang Yu
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8.,Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China, 410011
| | - Guangheng Zhu
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8
| | - Yi-Min She
- Centre for Biologics Research, Biologics and Genetic Therapies Directorate, HPFB, Health Canada, Ottawa, ON, Canada, K1A 0M2
| | - Terry Cyr
- Centre for Biologics Research, Biologics and Genetic Therapies Directorate, HPFB, Health Canada, Ottawa, ON, Canada, K1A 0M2
| | - Wenbin Fu
- Department of Acupuncture and Moxibustion, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, P.R. China, 510120.,Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P.R. China, 510000
| | - Guoqing Liu
- Institute of Cardiovascular Science, Peking University Health Science Center, Beijing, P.R. China, 100083
| | - Philip W Connelly
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada, M5S 1A1.,Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8
| | - Margaret L Rand
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada, M5S 1A1.,Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, ON, Canada, M5G 1X8
| | - Khosrow Adeli
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada, M5S 1A1.,Program in Molecular Structure & Function, The Hospital for Sick Children, Toronto, ON, Canada, M5G 1X8
| | - John Freedman
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada, M5S 1A1.,Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8.,Department of Medicine, University of Toronto, Toronto, ON, Canada, M5S 1A1
| | - Jeffrey E Lee
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada, M5S 1A1
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA, 45219
| | - Patrizia Marchese
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92037
| | - W Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA, 45219
| | - Shaun P Jackson
- Heart Research Institute, and Charles Perkins Centre, The University of Sydney, Camperdown, Australia, 2006.,Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92037
| | - Cheng Zhu
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA, 30332.,Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA, 30332.,Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA, 30332
| | - Zaverio M Ruggeri
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92037
| | - Heyu Ni
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada, M5S 1A1. .,Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, Canada, M5B 1W8. .,Canadian Blood Services Centre for Innovation, Toronto, ON, Canada, M5G 2M1. .,Department of Medicine, University of Toronto, Toronto, ON, Canada, M5S 1A1. .,Department of Physiology, University of Toronto, Toronto, ON, Canada, M5S 1A1.
| |
Collapse
|
123
|
Kim TH, Oh DK, Oh YM, Lee SW, Do Lee S, Lee JS. Fibrinogen as a potential biomarker for clinical phenotype in patients with chronic obstructive pulmonary disease. J Thorac Dis 2018; 10:5260-5268. [PMID: 30416773 DOI: 10.21037/jtd.2018.08.52] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Under the Food and Drug Administration's drug development tool qualification program, fibrinogen is the first biomarker drug development tool used in chronic obstructive pulmonary disease (COPD). However, the correlation between fibrinogen and exacerbations among Korean patients with COPD remains unclear. Methods In this retrospective cross-sectional study, we included patients with COPD for whom plasma fibrinogen assessment results, without exacerbation, were available. Then, we compared subgroups according to fibrinogen level (threshold: 350 mg/dL). We used multivariate linear regression analysis to investigate the clinical phenotype of COPD with high fibrinogen level, analyzed the correlation between the COPD severity indexes and fibrinogen level. Results Of 140 patients, we confirmed 48 (34.3%) patients in the high-level fibrinogen group. The high-level group demonstrated a medical history of more exacerbations than the low-level group. Lung functions [forced expiratory volume in 1 s (FEV1), forced vital capacity, and 6-minute walk distance] were more deteriorated in the high-level group. Multivariate regression analysis revealed that fibrinogen level was associated with high COPD assessment test score, and experience of exacerbation. Fibrinogen level exhibited a statistically significant positive correlation with COPD severity indexes. Conclusions High fibrinogen level seems to reflect frequent exacerbation and severe symptomatic phenotypes in Korean patients with COPD.
Collapse
Affiliation(s)
- Tae Hoon Kim
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Dong Kyu Oh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yeon-Mok Oh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sei Won Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang Do Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Seung Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
124
|
Alqahtani SA, Alhawiti NM. Administration of testosterone improves the prothrombotic and antifibrinolytic parameters associated with its deficiency in an orchidectiomized rat model. Platelets 2018; 30:624-630. [DOI: 10.1080/09537104.2018.1499886] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Sultan A. Alqahtani
- College of Medicine, King Saud bin Abdulaziz University for Health Science (KSAU-HS), Riyadh, Kingdom of Saudi Arabia (KSA)
| | - Naif M. Alhawiti
- College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Kingdom of Saudi Arabia (KSA)
| |
Collapse
|
125
|
Zhang Y, Kim DK, Jung YS, Kim YH, Lee YS, Kim J, Jeong WI, Lee IK, Cho SJ, Dooley S, Lee CH, Choi HS. Inverse agonist of ERRγ reduces cannabinoid receptor type 1-mediated induction of fibrinogen synthesis in mice with a high-fat diet-intoxicated liver. Arch Toxicol 2018; 92:2885-2896. [PMID: 30019168 DOI: 10.1007/s00204-018-2270-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/12/2018] [Indexed: 12/12/2022]
Abstract
Upon liver intoxication with malnutrition or high-fat diet feeding, fibrinogen is synthesized by hepatocytes and secreted into the blood in human and mouse. Its primary function is to occlude blood vessels upon damage and thereby stop excessive bleeding. High fibrinogen levels may contribute to the development of pathological thrombosis, which is one mechanism linking fatty liver disease with cardiovascular disease. Our previous results present ERRγ as key regulator of hepatocytic fibrinogen gene expression in human. In a therapeutic approach, we now tested ERRγ inverse agonist GSK5182 as regulator of fibrinogen levels in mouse hyperfibrinogenemia caused by diet-induced obesity and in mouse hepatocytes. ACEA, a CB1R agonist, up-regulated transcription of mouse fibrinogen via induction of ERRγ, whereas knockdown of ERRγ attenuated the effect of ACEA (10 µM) on fibrinogen expression in AML12 mouse hepatocytes. Deletion analyses of the mouse fibrinogen γ (FGG) gene promoter and ChIP assays revealed binding sites for ERRγ on the mouse FGG promoter. ACEA or adenovirus ERRγ injection induced FGA, FGB and FGG mRNA and protein expression in mouse liver, while ERRγ knockdown with Ad-shERRγ attenuated ACEA-mediated induction of fibrinogen gene expression. Moreover, mice maintained on a high-fat diet (HFD) expressed higher levels of fibrinogen, whereas cannabinoid receptor type 1 (CB1R)-KO mice fed an HFD had nearly normal fibrinogen levels. Finally, GSK5182 (40 mg/kg) strongly inhibits the ACEA (10 mg/kg) or HFD-mediated induction of fibrinogen level in mice. Taken together, targeting ERRγ with its inverse agonist GSK5182 represents a promising therapeutic strategy for ameliorating hyperfibrinogenemia.
Collapse
Affiliation(s)
- Yaochen Zhang
- National Creative Research Initiatives Center for Nuclear Receptor Signals, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Don-Kyu Kim
- National Creative Research Initiatives Center for Nuclear Receptor Signals, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Yoon Seok Jung
- National Creative Research Initiatives Center for Nuclear Receptor Signals, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Yong-Hoon Kim
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Yong Soo Lee
- National Creative Research Initiatives Center for Nuclear Receptor Signals, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Jina Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Won-Il Jeong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea.,Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Sung Jin Cho
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea.,New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Steven Dooley
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Chul-Ho Lee
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Hueng-Sik Choi
- National Creative Research Initiatives Center for Nuclear Receptor Signals, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea.
| |
Collapse
|
126
|
Nersesian PV, Han HR, Yenokyan G, Blumenthal RS, Nolan MT, Hladek MD, Szanton SL. Loneliness in middle age and biomarkers of systemic inflammation: Findings from Midlife in the United States. Soc Sci Med 2018; 209:174-181. [PMID: 29735350 PMCID: PMC6013269 DOI: 10.1016/j.socscimed.2018.04.007] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 03/15/2018] [Accepted: 04/05/2018] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Middle-aged adults who are lonely have an elevated likelihood of death. Systemic inflammation may contribute to these increased odds. Using population-level data, this study tested if systemic inflammation is associated with loneliness in a broad age range of middle-aged adults in the United States. METHODS This study used data from the Midlife in the US (MIDUS) survey Biomarker Project, which collected data on psychological, social, and physiological measures from a sample of middle-aged adults. This sample included the 927 participants who were 35-64 years at Biomarker Project data collection. MIDUS collected baseline data from 1995-1996 and a follow-up survey was conducted from 2004-2006. The baseline Milwaukee sample of African Americans was collected in 2005-2006 and the biomarker database was collected in 2004-2009. Biomarkers were obtained from a fasting blood sample. Self-reported loneliness was categorized as feeling lonely or not feeling lonely. Hierarchical regressions examined the association between biomarkers of systemic inflammation (interleukin-6, fibrinogen, C-reactive protein) and feeling lonely, adjusted for covariates. RESULTS Twenty-nine percent of the sample reported feeling lonely most or some of the time. There was a positive significant relationship between loneliness and the three systemic inflammation biomarkers after controlling for covariates: interleukin-6 (n = 873) (b [se] = 0.07 [0.03], p = .014); fibrinogen (n = 867) (b [se] = 18.24 [7.12], p = .011); and C-reactive protein (n = 867) (b [se] = 0.08 [0.04], p = .035). CONCLUSIONS Feeling lonely is associated with systemic inflammation in middle-aged community-dwelling US adults.
Collapse
Affiliation(s)
- Paula V Nersesian
- Johns Hopkins School of Nursing, 525 N. Wolfe St., Baltimore, MD 21205, USA.
| | - Hae-Ra Han
- Johns Hopkins School of Nursing, 525 N. Wolfe St., Baltimore, MD 21205, USA.
| | - Gayane Yenokyan
- Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD 21205, USA.
| | - Roger S Blumenthal
- Johns Hopkins School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA.
| | - Marie T Nolan
- Johns Hopkins School of Nursing, 525 N. Wolfe St., Baltimore, MD 21205, USA.
| | - Melissa D Hladek
- Johns Hopkins School of Nursing, 525 N. Wolfe St., Baltimore, MD 21205, USA.
| | - Sarah L Szanton
- Johns Hopkins School of Nursing, 525 N. Wolfe St., Baltimore, MD 21205, USA; Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD 21205, USA.
| |
Collapse
|
127
|
Fisk M, Cheriyan J, Mohan D, McEniery CM, Forman J, Cockcroft JR, Rudd JHF, Tal-Singer R, Hopkinson NS, Polkey MI, Wilkinson IB. Vascular inflammation and aortic stiffness: potential mechanisms of increased vascular risk in chronic obstructive pulmonary disease. Respir Res 2018; 19:100. [PMID: 29793484 PMCID: PMC5968523 DOI: 10.1186/s12931-018-0792-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 04/27/2018] [Indexed: 01/09/2023] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a complex inflammatory condition in which an important extra-pulmonary manifestation is cardiovascular disease. We hypothesized that COPD patients would have increased aortic inflammation and stiffness, as candidate mechanisms mediating increased cardiovascular risk, compared to two negative control groups: healthy never-smokers and smokers without COPD. We also studied patients with COPD due to alpha− 1 antitrypsin deficiency (α1ATD) as a comparator lung disease group. Methods Participants underwent 18F-Fluorodeoxyglucose (FDG) positron emission tomography imaging to quantify aortic inflammation as the tissue-to-blood-ratio (TBR) of FDG uptake. Aortic stiffness was measured by carotid-femoral aortic pulse wave velocity (aPWV). Results Eighty-five usual COPD (COPD due to smoking), 12 α1ATD-COPD patients and 12 each smokers and never-smokers were studied. There was no difference in pack years smoked between COPD patients and smokers (45 ± 25 vs 37 ± 19, p = 0.36), but α1ATD patients smoked significantly less (19 ± 11, p < 0.001 for both). By design, spirometry measures were lower in COPD and α1ATD-COPD patients compared to smokers and never-smokers. Aortic inflammation and stiffness were increased in COPD (TBR: 1.90 ± 0.38, aPWV: 9.9 ± 2.6 m/s) and α1ATD patients (TBR: 1.94 ± 0.43, aPWV: 9.5 ± 1.8 m/s) compared with smokers (TBR: 1.74 ± 0.30, aPWV: 7.8 ± 1.8 m/s, p < 0.05 all) and never-smokers (TBR: 1.71 ± 0.34, aPWV: 7.9 ± 1.7 m/s, p ≤ 0.05 all). Conclusions In this cross-sectional prospective study, novel findings were that both usual COPD and α1ATD-COPD patients have increased aortic inflammation and stiffness compared to smoking and never-smoking controls, regardless of smoking history. These findings suggest that the presence of COPD lung disease per se may be associated with adverse aortic wall changes, and aortic inflammation and stiffening are potential mechanisms mediating increased vascular risk observed in COPD patients. Electronic supplementary material The online version of this article (10.1186/s12931-018-0792-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marie Fisk
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK.
| | - Joseph Cheriyan
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK.,Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Divya Mohan
- NIHR Respiratory Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust and Imperial College, London, UK.,GSK R&D, King of Prussia, Pennsylvania, USA
| | - Carmel M McEniery
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Julia Forman
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - John R Cockcroft
- Department of Cardiology, Wales Heart Research Institute, Cardiff University, Cardiff, UK
| | - James H F Rudd
- Division of Cardiovascular Medicine, University of Cambridge & Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - Nicholas S Hopkinson
- NIHR Respiratory Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust and Imperial College, London, UK
| | - Michael I Polkey
- NIHR Respiratory Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust and Imperial College, London, UK
| | - Ian B Wilkinson
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK.,Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
128
|
Yu E, Hsu HY, Huang CY, Hwang LC. Inflammatory Biomarkers and Risk of Atherosclerotic Cardiovascular Disease. Open Med (Wars) 2018; 13:208-213. [PMID: 29845121 PMCID: PMC5968412 DOI: 10.1515/med-2018-0032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 02/15/2018] [Indexed: 12/20/2022] Open
Abstract
Background Non-alcoholic fatty liver disease is an increasing health issue that associates with the development of atherosclerotic cardiovascular disease. This study correlates the association between fatty liver and inflammatory biomarkers with cardiovascular risk scores. Methodology This cross-sectional study enrolled 10,181 health examination participants from Northern Taiwan and administered a standardized questionnaire with important biochemical tests and abdominal sonography. To assess concentrations of inflammatory markers high sensitivity C-reactive protein (hs-CRP) and fibrinogen were used. Results Inflammatory marker levels were significantly increased with increasing fatty liver. In multivariate logistic regression analysis adjusted for major confounding factors, the odds ratios of elevated hs-CRP and fibrinogen were significantly higher in participants with mild or moderate-to-severe fatty liver compared to healthy individuals. The cardiovascular risk scores, above cut-off level 10%, were associated with higher levels of inflammatory biomarkers and fatty liver; odds ratio, 3.52 (2.60-4.77) for non-alcoholic fatty liver disease with hs-CRP, and 2.92 (2.12-4.00) for non-alcoholic fatty liver disease with fibrinogen. Conclusion Inflammatory biomarkers (hs-CRP and fibrinogen) are significantly associated with augmentation of fatty liver. Non-alcoholic fatty liver disease may be a predictor of future atherosclerotic cardiovascular disease, and the prediction value increases on adding inflammatory biomarkers levels.
Collapse
Affiliation(s)
- En Yu
- Department of Family Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Hsin-Yin Hsu
- Department of Family Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Chun-Yuan Huang
- Department of Family Medicine, China Medical University Hospital Taipei Branch, Taipei, Taiwan
| | - Lee-Ching Hwang
- Department of Family Medicine, Mackay Memorial Hospital, No. 92, Sec. 2, Zhongshan North Road, Taipei City 10449, Taiwan.,Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| |
Collapse
|
129
|
Ospemifene's effects on lipids and coagulation factors: a post hoc analysis of phase 2 and 3 clinical trial data. Menopause 2018; 24:1167-1174. [PMID: 28509812 PMCID: PMC5617371 DOI: 10.1097/gme.0000000000000900] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Objective: To evaluate the effect of ospemifene 60 mg on the lipid and coagulation parameters of postmenopausal women using data from five phase 2 and 3 clinical trials. Methods: Data for lipids and coagulation factors for 2,166 postmenopausal women were pooled from five randomized, placebo-controlled studies. Lipid and coagulation parameters included in this analysis were total cholesterol, high-density lipoproteins (HDL), low-density lipoproteins (LDL), triglycerides, activated partial thromboplastin time (aPTT), fibrinogen, antithrombin antigen, protein C Ag, and protein S Ag free. Results: Mean percent changes in HDL and LDL were significantly greater with ospemifene versus placebo at month 3 (HDL: 4.4% vs 0.2%; LDL: −5.2% vs 2.4%), month 6 (HDL: 5.1% vs 1.5%; LDL: −6.7% vs 2.4%), and month 12 (HDL: 2.3% vs −1.9%; LDL: −7.0% vs −2.1%; P < 0.05, for all comparisons). Ospemifene significantly reduced total cholesterol at 6 months (−1.8% vs 1.6%; P = 0.0345 versus placebo), and changes in triglycerides with ospemifene were similar to placebo at all three time points. In subgroup analyses based on age, body mass index, and baseline triglyceride level, ospemifene increased HDL and decreased LDL, but had no significant effect on total cholesterol and triglycerides relative to placebo. Ospemifene significantly improved fibrinogen and protein C antigen levels relative to placebo at months 3 (−8.7% vs −0.8% and −2.7% vs 0.5%, respectively), 6 (−6.0% vs 6.7% and −3.6 vs 8.0%), and 12 (−8.7% vs 7.3% and −4.5% vs 6.6%; P < 0.01, for all). The levels of all coagulation factors remained within the normal range throughout the studies. Conclusion: Ospemifene 60 mg does not have a detrimental effect on lipid and coagulation parameters of postmenopausal women with up to 12 months of use.
Collapse
|
130
|
Homeostasis model assessment of insulin resistance in relation to the poor functional outcomes in nondiabetic patients with ischemic stroke. Biosci Rep 2018; 38:BSR20180330. [PMID: 29588341 PMCID: PMC5938425 DOI: 10.1042/bsr20180330] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 03/24/2018] [Accepted: 03/26/2018] [Indexed: 12/11/2022] Open
Abstract
Whether insulin resistance (IR) predicts worse functional outcome in ischemic stroke is still a matter of debate. The aim of the present study is to determine the association between IR and risk of poor outcome in 173 Chinese nondiabetic patients with acute ischemic stroke. This is a prospective, population-based cohort study. Insulin sensitivity, expressed by the homeostasis model assessment (HOMA) of insulin sensitivity (HOMA index = (fasting insulin × fasting glucose)/22.5). IR was defined by HOMA-IR index in the top quartile (Q4). Functional impairment was evaluated at discharge using the modified Rankin scale (mRS). The median (interquartile range) HOMA-IR was 2.14 (1.17–2.83), and Q4 was at least 2.83. There was a significantly positive correlation between HOMA-IR and National Institutes of Health Stroke Scale (r = 0.408; P<0.001). In multivariate analyses, patients in IR group were associated with a higher risk of poor functional outcome (odds ratio (OR) = 3.23; 95% confidence interval (CI) = 1.75–5.08; P=0.001). In multivariate models comparing the third and fourth quartiles against the first quartile of the HOMA-IR, levels of HOMA-IR were associated with poor outcome, and the adjusted risk of poor outcome increased by 207% (OR = 3.05 (95% CI 1.70–4.89), P=0.006) and 429% (5.29 (3.05–9.80), P<0.001). In a receiver operating characteristic curve (ROC) analysis of poor outcome, the area under the curve (AUC) increased from 0.80 to 0.84 (95% CI: 0.79–0.88) by adding HOMA-IR to clinical examination variables (P=0.02). High HOMA-IR index is associated with a poor functional outcome in nondiabetic patients with acute ischemic stroke.
Collapse
|
131
|
Panova-Noeva M, Schulz A, Arnold N, Hermanns MI, Prochaska JH, Laubert-Reh D, Spronk HM, Blettner M, Beutel M, Pfeiffer N, Münzel T, Lackner KJ, Ten Cate H, Wild PS. Coagulation and inflammation in long-term cancer survivors: results from the adult population. J Thromb Haemost 2018; 16:699-708. [PMID: 29431889 DOI: 10.1111/jth.13975] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Indexed: 01/07/2023]
Abstract
Essentials The increase of cancer survival remains curtailed by cardiovascular mortality. We studied a large range of inflammatory and coagulation biomarkers in long-term cancer survivors. Cancer history has an important impact on mortality independent of cardiovascular risk factors. Fibrinogen and von Willebrand factor are potential biomarkers in survivors of increased mortality. SUMMARY Background The advances in cancer treatment and detection of early cancer have resulted in a steady increase in the number of of cancer survivors over the years. However, because of the long-term toxic effects of chemotherapy and radiotherapy, the incidence of cardiovascular disease (CVD) is increasing in survivors. Objectives To investigate traditional cardiovascular risk factors (CVRFs), inflammation and the coagulation profile in long-term cancer survivors (cancer diagnosis ≥ 5 years) from a large adult population-based study sample. Methods The presence of cardiovascular risk factors (CVRFs) and laboratory markers were compared in individuals with (n = 723) and without (n = 13626) a long-term history of cancer from the Gutenberg Health Study. Data on coagulation factors, D-dimer and von Willebrand factor (VWF) activity were available for 4974 individuals (n = 244 cancer survivors). Results In multivariable regression models, a history of cancer was, independently of CVRFs and CVD, associated with higher fibrinogen levels (β 6.99, 95% confidence interval [CI] 1.16-12.8), VWF activity (β 5.08, 95% CI 0.02-10.1), and antithrombin activity (β 1.85, 95% CI 0.44-3.27). Cancer survivors with CVD showed notably higher VWF activity than individuals with CVD without a history of cancer, with a difference in the means of 23.0 (7.9-38.1). Multivariate Cox regression analysis, adjusted for CVRFs, confirmed that a long-term history of cancer is associated with a 72% higher mortality. Increased mortality in cancer survivors was dependent on fibrinogen level and VWF activity level. Conclusion Cancer survivors showed a worse inflammation and coagulation profile than individuals without a history of cancer. Overall mortality in long-term cancer survivors was increased independently of traditional CVRFs. These results underline the need to further investigate plasma biomarkers as complementary cardiovascular risk predictors in cancer survivors.
Collapse
Affiliation(s)
- M Panova-Noeva
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - A Schulz
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - N Arnold
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - M I Hermanns
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - J H Prochaska
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site RhineMain, Mainz, Germany
| | - D Laubert-Reh
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - H M Spronk
- Laboratory for Clinical Thrombosis and Hemostasis, Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, the Netherlands
| | - M Blettner
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - M Beutel
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - N Pfeiffer
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - T Münzel
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site RhineMain, Mainz, Germany
- Center for Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - K J Lackner
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - H Ten Cate
- Laboratory for Clinical Thrombosis and Hemostasis, Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, the Netherlands
| | - P S Wild
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site RhineMain, Mainz, Germany
| |
Collapse
|
132
|
Olie RH, van der Meijden PE, ten Cate H. The coagulation system in atherothrombosis: Implications for new therapeutic strategies. Res Pract Thromb Haemost 2018; 2:188-198. [PMID: 30046721 PMCID: PMC6055505 DOI: 10.1002/rth2.12080] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 01/05/2018] [Indexed: 12/13/2022] Open
Abstract
Clinical manifestations of atherosclerotic disease include coronary artery disease (CAD), peripheral artery disease (PAD), and stroke. Although the role of platelets is well established, evidence is now accumulating on the contribution of coagulation proteins to the processes of atherosclerosis and atherothrombosis. Coagulation proteins not only play a role in fibrin formation and platelet activation, but also mediate various biological and pathophysiologic processes through activation of protease-activated-receptors (PARs). Thus far, secondary prevention in patients with CAD/PAD has been the domain of antiplatelet therapy, however, residual atherothrombotic risks remain substantial. Therefore, combining antiplatelet and anticoagulant therapy has gained more attention. Recently, net clinical benefit of combining aspirin with low-dose rivaroxaban in patients with stable atherosclerotic disease has been demonstrated. In this review, based on the State of the Art lecture "Clotting factors and atherothrombosis" presented at the ISTH Congress 2017, we highlight the role of coagulation proteins in the pathophysiology of atherothrombosis, and specifically focus on therapeutic strategies to decrease atherothrombotic events by optimization of vascular protection.
Collapse
Affiliation(s)
- Renske H. Olie
- Department of Internal MedicineMaastricht University Medical Center+ (MUMC+)MaastrichtThe Netherlands
- Thrombosis Expertise CenterMUMC+MaastrichtThe Netherlands
- Laboratory for Clinical Thrombosis and HemostasisMaastricht UniversityMaastrichtThe Netherlands
| | - Paola E.J. van der Meijden
- Thrombosis Expertise CenterMUMC+MaastrichtThe Netherlands
- Laboratory for Clinical Thrombosis and HemostasisMaastricht UniversityMaastrichtThe Netherlands
| | - Hugo ten Cate
- Department of Internal MedicineMaastricht University Medical Center+ (MUMC+)MaastrichtThe Netherlands
- Thrombosis Expertise CenterMUMC+MaastrichtThe Netherlands
- Laboratory for Clinical Thrombosis and HemostasisMaastricht UniversityMaastrichtThe Netherlands
- Center for Thrombosis and HaemostasisGutenberg UniversityMainzGermany
| |
Collapse
|
133
|
Egnot NS, Barinas-Mitchell E, Criqui MH, Allison MA, Ix JH, Jenny NS, Wassel CL. An exploratory factor analysis of inflammatory and coagulation markers associated with femoral artery atherosclerosis in the San Diego Population Study. Thromb Res 2018; 164:9-14. [PMID: 29459247 PMCID: PMC5899938 DOI: 10.1016/j.thromres.2018.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/24/2018] [Accepted: 02/08/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIMS Several biomarkers of inflammation and coagulation have been implicated in lower extremity atherosclerosis. We utilized an exploratory factor analysis (EFA) to identify distinct factors derived from circulating inflammatory and coagulation biomarkers then examined the associations of these factors with measures of lower extremity subclinical atherosclerosis, including the ankle-brachial index (ABI), common and superficial femoral intima-media thickness (IMT), and atherosclerotic plaque presence, burden, and characteristics. METHODS The San Diego Population Study (SDPS) is a prospective, community-living, multi-ethnic cohort of 1103 men and women averaged age 70. Regression analysis was used to assess cross-sectional associations between the identified groupings of biomarkers (factors) and the ABI and femoral artery atherosclerosis measurements. RESULTS Two biomarker factors emerged from the factor analysis. Factor 1 consisting of C-reactive protein (CRP), interleukin (IL)-6, and fibrinogen was significantly associated with higher odds (OR = 1.99, p < 0.01) of a borderline ABI value (0.91-0.99), while Factor 2 containing D-dimer and pentraxin (PTX)-3 was significantly associated with higher common femoral artery (CFA) IMT (β = 0.23, p < 0.01) and lower ABI (β = -0.03, p < 0.01). CONCLUSIONS Two groupings of biomarkers were identified via EFA of seven circulating biomarkers of inflammation and coagulation. These distinct groups are differentially associated with markers of lower extremity subclinical atherosclerosis. Our findings suggest that high inflammatory and coagulation burden were better markers of more severe lower-extremity disease as indicated by low ABI rather than early atherosclerotic lesion development in the femoral artery.
Collapse
Affiliation(s)
- Natalie Suder Egnot
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States.
| | - Emma Barinas-Mitchell
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael H Criqui
- Division of Preventive Medicine, Department of Family Medicine and Public Health, School of Medicine, University of California-San Diego, La Jolla, CA, United States
| | - Matthew A Allison
- Division of Preventive Medicine, Department of Family Medicine and Public Health, School of Medicine, University of California-San Diego, La Jolla, CA, United States
| | - Joachim H Ix
- Division of Preventive Medicine, Department of Family Medicine and Public Health, School of Medicine, University of California-San Diego, La Jolla, CA, United States; Division of Nephrology, Department of Medicine, School of Medicine, University of California-San Diego, La Jolla, CA, United States
| | - Nancy S Jenny
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Christina L Wassel
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| |
Collapse
|
134
|
Fisk M, Cheriyan J, Mohan D, Forman J, Mäki-Petäjä KM, McEniery CM, Fuld J, Rudd JHF, Hopkinson NS, Lomas DA, Cockcroft JR, Tal-Singer R, Polkey MI, Wilkinson IB. The p38 mitogen activated protein kinase inhibitor losmapimod in chronic obstructive pulmonary disease patients with systemic inflammation, stratified by fibrinogen: A randomised double-blind placebo-controlled trial. PLoS One 2018; 13:e0194197. [PMID: 29566026 PMCID: PMC5863984 DOI: 10.1371/journal.pone.0194197] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 02/16/2018] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Cardiovascular disease is a major cause of morbidity and mortality in COPD patients. Systemic inflammation associated with COPD, is often hypothesised as a causal factor. p38 mitogen-activated protein kinases play a key role in the inflammatory pathogenesis of COPD and atherosclerosis. OBJECTIVES This study sought to evaluate the effects of losmapimod, a p38 mitogen-activated protein kinase (MAPK) inhibitor, on vascular inflammation and endothelial function in chronic obstructive pulmonary disease (COPD) patients with systemic inflammation (defined by plasma fibrinogen >2·8g/l). METHODS This was a randomised, double-blind, placebo-controlled, Phase II trial that recruited COPD patients with plasma fibrinogen >2.8g/l. Participants were randomly assigned by an online program to losmapimod 7·5mg or placebo tablets twice daily for 16 weeks. Pre- and post-dose 18F-Fluorodeoxyglucose positron emission tomography co-registered with computed tomography (FDG PET/CT) imaging of the aorta and carotid arteries was performed to quantify arterial inflammation, defined by the tissue-to-blood ratio (TBR) from scan images. Endothelial function was assessed by brachial artery flow-mediated dilatation (FMD). RESULTS We screened 160 patients, of whom, 36 and 37 were randomised to losmapimod or placebo. The treatment effect of losmapimod compared to placebo was not significant, at -0·05 for TBR (95% CI: -0·17, 0·07), p = 0·42, and +0·40% for FMD (95% CI: -1·66, 2·47), p = 0·70. The frequency of adverse events reported was similar in both treatment groups. CONCLUSIONS In this plasma fibrinogen-enriched study, losmapimod had no effect on arterial inflammation and endothelial function at 16 weeks of treatment, although it was well tolerated with no significant safety concerns. These findings do not support the concept that losmapimod is an effective treatment for the adverse cardiovascular manifestations of COPD.
Collapse
Affiliation(s)
- Marie Fisk
- Department of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom
| | - Joseph Cheriyan
- Department of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Divya Mohan
- NIHR Respiratory Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust and Imperial College, London, United Kingdom
- GSK R&D, King of Prussia, Pennsylvania, United States of America
| | - Julia Forman
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Kaisa M. Mäki-Petäjä
- Department of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom
| | - Carmel M. McEniery
- Department of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom
| | - Jonathan Fuld
- Department of Respiratory Medicine, University of Cambridge & Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - James H. F. Rudd
- Department of Cardiovascular Medicine, University of Cambridge & Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Nicholas S. Hopkinson
- NIHR Respiratory Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust and Imperial College, London, United Kingdom
| | - David A. Lomas
- Department of UCL Respiratory, Division of Medicine, Rayne Building, University College London, London, United Kingdom
| | - John R. Cockcroft
- Department of Cardiology, Wales Heart Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Ruth Tal-Singer
- GSK R&D, King of Prussia, Pennsylvania, United States of America
| | - Michael I. Polkey
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Ian B. Wilkinson
- Department of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
135
|
Hassan M. CANTOS: A breakthrough that proves the inflammatory hypothesis of atherosclerosis. Glob Cardiol Sci Pract 2018; 2018:2. [PMID: 29644229 PMCID: PMC5857062 DOI: 10.21542/gcsp.2018.2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Atherosclerosis is no longer considered solely a disorder of subintimal deposition of modified low-density lipoprotein particles in the arterial wall. Rather, it is known to be a chronic inflammatory disorder. No evidence has shown that reducing vascular inflammation in the absence of concomitant lowering of lipoproteins levels reduces the rates of adverse cardiovascular (CV) events. Canakinumab, a fully human monoclonal antibody that neutralizes interleukin (IL)-1β, significantly reduced the rate of recurrent CV events in patients with prior myocardial infarction in the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS). Canakinumab has no effect on CV or all-cause mortality, however it was associated with high incidence of fatal infections. Thus, the net benefit needs to be properly addressed in future studies that evaluate the potential benefit of the anti-inflammatory therapies and whether it can change clinical practice in the near future.
Collapse
Affiliation(s)
- Mohamed Hassan
- Cardiology Department, Cairo University, Cairo, Egypt.,Cardiology Department, Aswan Heart Centre, Aswan, Egypt
| |
Collapse
|
136
|
Lai CL, Xing JP, Liu XH, Qi J, Zhao JQ, Ji YR, Yang WX, Yan PJ, Luo CY, Ruan LF. Relationships of Inflammatory Factors and Risk Factors with Different Target Organ Damage in Essential Hypertension Patients. Chin Med J (Engl) 2018; 130:1296-1302. [PMID: 28524828 PMCID: PMC5455038 DOI: 10.4103/0366-6999.206343] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Atherosclerosis (AS) is an inflammatory disease. Inflammation was considered to play a role in the whole process of AS. This study aimed to analyze the relationships of inflammatory factors and risk factors with different target organ damages (TOD) in essential hypertension (EH) patients and to explore its clinical significance. METHODS A total of 294 EH patients were selected and divided into four groups according to their conditions of TOD. Forty-eight healthy subjects were selected as control. The clinical biochemical parameters, serum amyloid A, serum tryptase, and lipoprotein-associated phospholipase A2 (Lp-PLA2) in each group were detected, and the related risk factors were also statistically analyzed. RESULTS Fibrinogen (Fbg) was the most significant independent risk factor in acute coronary syndrome (ACS) group (odds ratio [OR]: 22.242, 95% confidence interval [CI]: 6.458-76.609, P< 0.001) with the largest absolute value of the standardized partial regression coefficient B' (b': 1.079). Lp-PLA2 was the most significant independent risk factor in stroke group (OR: 13.699, 95% CI: 5.236-35.837, P< 0.001) with b' = 0.708. Uric acid (UA) was the most significant independent risk factor in renal damage group (OR: 15.307, 95% CI: 4.022-58.250, P< 0.001) with b' = 1.026. CONCLUSIONS Fbg, Lp-PLA2, and UA are the strongest independent risk factors toward the occurrence of ACS, ischemic stroke, and renal damage in EH patients, thus exhibiting the greatest impacts on the occurrence of ACS, ischemic stroke, and renal damage in EH patients, respectively.
Collapse
Affiliation(s)
- Chun-Lin Lai
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Jin-Ping Xing
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Xiao-Hong Liu
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Jie Qi
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Jian-Qiang Zhao
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - You-Rui Ji
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Wu-Xiao Yang
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Pu-Juan Yan
- Postgraduate School of Shanxi Medical University, Taiyuan, Shanxi 030012, China
| | - Chun-Yan Luo
- Postgraduate School of Shanxi Medical University, Taiyuan, Shanxi 030012, China
| | - Lu-Fang Ruan
- Postgraduate School of Shanxi Medical University, Taiyuan, Shanxi 030012, China
| |
Collapse
|
137
|
Ebbeling CB, Klein GL, Luoto PK, Wong JMW, Bielak L, Eddy RG, Steltz SK, Devlin C, Sandman M, Hron B, Shimy K, Heymsfield SB, Wolfe RR, Wong WW, Feldman HA, Ludwig DS. A randomized study of dietary composition during weight-loss maintenance: Rationale, study design, intervention, and assessment. Contemp Clin Trials 2018; 65:76-86. [PMID: 29233719 PMCID: PMC6055230 DOI: 10.1016/j.cct.2017.12.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 12/07/2017] [Accepted: 12/09/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND While many people with overweight or obesity can lose weight temporarily, most have difficulty maintaining weight loss over the long term. Studies of dietary composition typically focus on weight loss, rather than weight-loss maintenance, and rely on nutrition education and dietary counseling, rather than controlled feeding protocols. Variation in initial weight loss and insufficient differentiation among treatments confound interpretation of results and compromise conclusions regarding the weight-independent effects of dietary composition. The aim of the present study was to evaluate three test diets differing in carbohydrate-to-fat ratio during weight-loss maintenance. DESIGN AND DIETARY INTERVENTIONS Following weight loss corresponding to 12±2% of baseline body weight on a standard run-in diet, 164 participants aged 18 to 65years were randomly assigned to one of three test diets for weight-loss maintenance through 20weeks (test phase). We fed them high-carbohydrate (60% of energy from carbohydrate, 20% fat), moderate-carbohydrate (40% carbohydrate, 40% fat), and low-carbohydrate (20% carbohydrate, 60% fat) diets, controlled for protein content (20% of energy). During a 2-week ad libitum feeding phase following the test phase, we assessed the effect of the test diets on body weight. OUTCOMES The primary outcome was total energy expenditure, assessed by doubly-labeled water methodology. Secondary outcomes included resting energy expenditure and physical activity, chronic disease risk factors, and variables to inform an understanding of physiological mechanisms by which dietary carbohydrate-to-fat ratio might influence metabolism. Weight change during the ad libitum feeding phase was conceptualized as a proxy measure of hunger.
Collapse
Affiliation(s)
- Cara B Ebbeling
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States.
| | - Gloria L Klein
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Patricia K Luoto
- Department of Food and Nutrition, Framingham State University, 100 State Street, PO Box 9101, Framingham, MA 01701, United States
| | - Julia M W Wong
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Lisa Bielak
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Ralph G Eddy
- Sodexo Inc., Framingham State University, 100 State Street, PO Box 9101, Framingham, MA 01701, United States
| | - Sarah K Steltz
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Courtenay Devlin
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Megan Sandman
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Bridget Hron
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States; Division of Gastroenterology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Kim Shimy
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Steven B Heymsfield
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| | - Robert R Wolfe
- University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - William W Wong
- Baylor College of Medicine, USDA/ARS Children's Nutrition Research Center, 1100 Bates Street, Houston, TX 77030, United States
| | - Henry A Feldman
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - David S Ludwig
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| |
Collapse
|
138
|
Robertson S, Miller MR. Ambient air pollution and thrombosis. Part Fibre Toxicol 2018; 15:1. [PMID: 29298690 PMCID: PMC5753450 DOI: 10.1186/s12989-017-0237-x] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/15/2017] [Indexed: 02/07/2023] Open
Abstract
Air pollution is a growing public health concern of global significance. Acute and chronic exposure is known to impair cardiovascular function, exacerbate disease and increase cardiovascular mortality. Several plausible biological mechanisms have been proposed for these associations, however, at present, the pathways are incomplete. A seminal review by the American Heart Association (2010) concluded that the thrombotic effects of particulate air pollution likely contributed to their effects on cardiovascular mortality and morbidity. The aim of the current review is to appraise the newly accumulated scientific evidence (2009-2016) on contribution of haemostasis and thrombosis towards cardiovascular disease induced by exposure to both particulate and gaseous pollutants.Seventy four publications were reviewed in-depth. The weight of evidence suggests that acute exposure to fine particulate matter (PM2.5) induces a shift in the haemostatic balance towards a pro-thrombotic/pro-coagulative state. Insufficient data was available to ascertain if a similar relationship exists for gaseous pollutants, and very few studies have addressed long-term exposure to ambient air pollution. Platelet activation, oxidative stress, interplay between interleukin-6 and tissue factor, all appear to be potentially important mechanisms in pollution-mediated thrombosis, together with an emerging role for circulating microvesicles and epigenetic changes.Overall, the recent literature supports, and arguably strengthens, the contention that air pollution contributes to cardiovascular morbidity by promoting haemostasis. The volume and diversity of the evidence highlights the complexity of the pathophysiologic mechanisms by which air pollution promotes thrombosis; multiple pathways are plausible and it is most likely they act in concert. Future research should address the role gaseous pollutants play in the cardiovascular effects of air pollution mixture and direct comparison of potentially susceptible groups to healthy individuals.
Collapse
Affiliation(s)
- Sarah Robertson
- Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0RQ, UK.
| | - Mark R Miller
- University/BHF Centre of Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
139
|
Gram AS, Petersen MB, Quist JS, Rosenkilde M, Stallknecht B, Bladbjerg EM. Effects of 6 Months of Active Commuting and Leisure-Time Exercise on Fibrin Turnover in Sedentary Individuals with Overweight and Obesity: A Randomised Controlled Trial. J Obes 2018; 2018:7140754. [PMID: 29887998 PMCID: PMC5977059 DOI: 10.1155/2018/7140754] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 03/05/2018] [Accepted: 04/15/2018] [Indexed: 12/11/2022] Open
Abstract
Obesity and exercise constitute important factors for cardiovascular disease risk, but the long-term effects of different exercise modalities on haemostatic biomarkers are not well elucidated. We investigated the effects of 6 months of active commuting or leisure-time exercise on measures of fibrin turnover in individuals who are overweight and obese. Ninety younger (20-40 years), sedentary, healthy women and men who are overweight and obese (BMI: 25-35 kg/m2) were randomised to 6 months of habitual lifestyle (CON, n=16), active commuting (BIKE, n=19), or leisure-time exercise of moderate (MOD, ∼50% VO2peak reserve, n=31) or vigorous intensity (VIG, ∼70% VO2peak reserve, n=24). Fasting blood samples (baseline and 3 and 6 months) were analysed for cholesterols and triglycerides, thrombin generation, prothrombin fragment 1 + 2, D-dimer, fibrin clot properties, and fibrinolytic activity. We observed no differences between CON, BIKE, MOD, and VIG during the intervention and no time effects for any of the variables measured despite increased VO2peak in all exercise groups. We found no difference between CON and all exercise groups combined and no gender-specific effects of exercise. Our findings suggest that thrombin generation capacity, coagulation activation, fibrin clot structure, and lysability are unaffected by long-term active commuting and leisure-time exercise in women and men who are overweight and obese.
Collapse
Affiliation(s)
- Anne Sofie Gram
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Martin Bæk Petersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas Salling Quist
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mads Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bente Stallknecht
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Else-Marie Bladbjerg
- Unit for Thrombosis Research, Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
- Department of Clinical Biochemistry, Hospital of South West Jutland, Esbjerg, Denmark
| |
Collapse
|
140
|
Loid P, Långström S, Viljakainen H, Mäkipernaa A, Heikinheimo M, Mäkitie O. Prothrombotic state in young females with severe early-onset obesity. Pediatr Res 2018; 83:2-4. [PMID: 29068434 DOI: 10.1038/pr.2017.248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Petra Loid
- University of Helsinki and Helsinki University Hospital, Children's Hospital, Helsinki, Finland
| | - Satu Långström
- University of Helsinki and Helsinki University Hospital, Children's Hospital, Helsinki, Finland
| | - Heli Viljakainen
- University of Helsinki and Helsinki University Hospital, Children's Hospital, Helsinki, Finland
- Folkhälsan Institute of Genetics and University of Helsinki, Helsinki, Finland
| | - Anne Mäkipernaa
- Children's Hospital and Hematology, Comprehensive Cancer Center, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Markku Heikinheimo
- University of Helsinki and Helsinki University Hospital, Children's Hospital, Helsinki, Finland
| | - Outi Mäkitie
- University of Helsinki and Helsinki University Hospital, Children's Hospital, Helsinki, Finland
- Folkhälsan Institute of Genetics and University of Helsinki, Helsinki, Finland
- Center for Molecular Medicine, Karolinska Institutet, and Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
141
|
Steptoe A, Kivimäki M, Lowe G, Rumley A, Hamer M. Blood Pressure and Fibrinogen Responses to Mental Stress as Predictors of Incident Hypertension over an 8-Year Period. Ann Behav Med 2017; 50:898-906. [PMID: 27401000 PMCID: PMC5126198 DOI: 10.1007/s12160-016-9817-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background Heightened blood pressure (BP) responses to mental stress predict raised BP levels over subsequent years, but evidence for associations with incident hypertension is limited, and the significance of inflammatory responses is uncertain. Purpose We investigated the relationship between BP and plasma fibrinogen responses to stress and incident hypertension over an average 8-year follow-up. Method Participants were 636 men and women (mean age 59.1 years) from the Whitehall II epidemiological cohort with no history of cardiovascular disease and hypertension. They performed standardized behavioral tasks (color/word conflict and mirror tracing), and hypertension was defined by clinic measures and medication status. Results Of participants in the highest systolic BP reactivity tertile, 29.3 % became hypertensive over the follow-up period compared with 16.5 % of those in the lowest tertile, with an odds ratio of 2.02 (95 % CI 1.17–3.88, p = 0.012) after adjustment for age, sex, grade of employment, body mass index, smoking, alcohol consumption, physical activity, follow-up time, subjective stress response, perceived task difficulty, perceived task engagement, and baseline BP. Similar associations were observed for diastolic BP reactivity (odds ratio 2.05, 95 % CI 1.23–3.40, p = 0.006) and for impaired systolic BP post-stress recovery (odds ratio 2.06, 95 % CI 1.19–3.57, p = 0.010). Fibrinogen reactions to tasks also predicted future hypertension in women (odds ratio 2.64, 95 % CI 1.11–6.30, p = 0.029) but not men. Conclusions These data suggest that heightened cardiovascular and inflammatory reactivity to mental stress is associated with hypertension risk, and may be a mechanism through which psychosocial factors impact on the development of hypertension.
Collapse
Affiliation(s)
- Andrew Steptoe
- Department of Epidemiology and Public Health, University College London, 1-19 Torrington Place, London, WC1E 6BT, UK.
| | - Mika Kivimäki
- Department of Epidemiology and Public Health, University College London, 1-19 Torrington Place, London, WC1E 6BT, UK
| | - Gordon Lowe
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland
| | - Ann Rumley
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland
| | - Mark Hamer
- National Centre for Sport & Exercise Medicine, Loughborough University, Loughborough, UK
| |
Collapse
|
142
|
Paraboschi EM, Duga S, Asselta R. Fibrinogen as a Pleiotropic Protein Causing Human Diseases: The Mutational Burden of Aα, Bβ, and γ Chains. Int J Mol Sci 2017; 18:E2711. [PMID: 29240685 PMCID: PMC5751312 DOI: 10.3390/ijms18122711] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/11/2017] [Accepted: 12/13/2017] [Indexed: 12/19/2022] Open
Abstract
Fibrinogen is a highly pleiotropic protein that is involved in the final step of the coagulation cascade, wound healing, inflammation, and angiogenesis. Heterozygous mutations in Aα, Bβ, or γ fibrinogen-chain genes (FGA, FGB, FGG) have been described as being responsible for fibrinogen deficiencies (hypofibrinogenemia, hypo-dysfibrinogenemia, dysfibrinogenemia) and for more rare conditions, such as fibrinogen storage disease and hereditary renal amyloidosis. Instead, biallelic mutations have been associated with afibrinogenemia/severe hypofibrinogenemia, i.e., the severest forms of fibrinogen deficiency, affecting approximately 1-2 cases per million people. However, the "true" prevalence for these conditions on a global scale is currently not available. Here, we defined the mutational burden of the FGA, FGB, and FGG genes, and estimated the prevalence of inherited fibrinogen disorders through a systematic analysis of exome/genome data from ~140,000 individuals belonging to the genome Aggregation Database. Our analysis showed that the world-wide prevalence for recessively-inherited fibrinogen deficiencies could be 10-fold higher than that reported so far (prevalence rates vary from 1 in 10⁶ in East Asians to 24.5 in 10⁶ in non-Finnish Europeans). The global prevalence for autosomal-dominant fibrinogen disorders was estimated to be ~11 in 1000 individuals, with heterozygous carriers present at a frequency varying from 3 every 1000 individuals in Finns, to 1-2 every 100 individuals among non-Finnish Europeans and Africans/African Americans. Our analysis also allowed for the identification of recurrent (i.e., FGG-p.Ala108Gly, FGG-Thr47Ile) or ethnic-specific mutations (e.g., FGB-p.Gly103Arg in Admixed Americans, FGG-p.Ser245Phe in Africans/African Americans).
Collapse
Affiliation(s)
- Elvezia Maria Paraboschi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy.
| | - Stefano Duga
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy.
- Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano, Milan, Italy.
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy.
- Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano, Milan, Italy.
| |
Collapse
|
143
|
Morange PE, Alessi MC. Thrombosis in central obesity and metabolic syndrome: Mechanisms and epidemiology. Thromb Haemost 2017; 110:669-80. [DOI: 10.1160/th13-01-0075] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 04/20/2013] [Indexed: 12/19/2022]
Abstract
summaryCentral obesity is a key feature of the metabolic syndrome (metS), a multiplex risk factor for subsequent development of type 2 diabetes and cardiovascular disease. Many metabolic alterations closely related to this condition exert effects on platelets and vascular cells. A procoagulant and hypofibrinolytic state has been identified, mainly underlain by inflammation, oxidative stress, dyslipidaemia, and ectopic fat that accompany central obesity. In support of these data, central obesity independently predisposes not only to atherothrombosis but also to venous thrombosis.
Collapse
|
144
|
Baumert J, Karakas M, Greven S, Rückerl R, Peters A, Koenig W. Variability of fibrinogen measurements in post-myocardial infarction patients. Thromb Haemost 2017; 107:895-902. [DOI: 10.1160/th11-10-0703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 01/18/2012] [Indexed: 11/05/2022]
Abstract
SummaryElevated fibrinogen levels are strongly and consistently associated with incident coronary heart disease (CHD). A possible causal contribution of fibrinogen in the pathway leading to atherothrombotic cardiovascular disease complications has been suggested. However, for implementation in clinical practice, data on validity and reliability, which are still scarce, are needed that are still scarce, especially in subjects with a history of CHD. For the present study, levels of plasma fibrinogen were measured in 200 post-myocardial infarction (post-MI) patients aged 39–76 years, with approximately six blood samples collected at monthly intervals between May 2003 and March 2004, giving a total of 1,144 samples. Inter-individual variability (between-subject variance component, VCb and coefficient of variation, CVb), intra-individual and analytical variability (VCw+a and CVw+a), intraclass correlation coefficient (ICC) and the number of measurements required for an ICC of 0.75 were estimated to assess the reliability of serial fibrinogen measurements. Mean fibrinogen concentration of all subjects over all samples was 3.34 g/l (standard deviation 0.67). Between-subject variation for fibrinogen was VCb = 0.34 (CVb,=17.5%) whereas within-subject and analytical variation was estimated as VCw+a = 0.14 (CVw+a=11.0%). The variation was mainly explained by between-subject variability, shown by the proportion of total variance of 71.3%. Two different measurements were required to reach sufficient reliability, if subjects with extreme values were not excluded. The present study indicates a fairly good reproducibility of serial individual fibrinogen measurements in post-MI subjects.
Collapse
|
145
|
Tiedje V, Dunkler D, Ay C, Horvath B, Quehenberger P, Pabinger M, Zielinski C, Mannhalter C, Pabinger I. The role of fibrinogen plasma levels, the –455G>A fibrinogen and the factor XIII A subunit (FXIII-A) Val34Leu polymorphism in cancer-associated venous thrombosis. Thromb Haemost 2017; 106:908-13. [DOI: 10.1160/th11-04-0278] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 07/22/2011] [Indexed: 11/05/2022]
Abstract
SummaryVenous thromboembolism (VTE) is a life-threatening complication in cancer patients. Identification of risk factors has been in focus in the past years. Functional single nucleotide polymorphisms (SNP) of coagulation factors known to influence the concentration or function may be considered to influence the risk of VTE in cancer patients. We evaluated the influence of fibrinogen plasma levels, the –455G>A SNP in the fibrinogen beta gene and the Val34Leu (163G>T) SNP in the factor XIII A-subunit (FXIII-A) gene on the risk of VTE. In 1,079 tumour patients recruited for the prospective Vienna Cancer and Thrombosis Study (CATS) fibrinogen levels were determined by the Clauss method. The FXIII-A Val34Leu and the fibrinogen –455G>A SNPs were tested by allele-specific PCR. The median follow-up time was 604 days, 83 thrombotic events occurred. The median fibrinogen level was 381 mg/dl (25th-75th percentile: 312 to 467). In a multivariable Cox model adjusted to chemotherapy, surgery, radiotherapy, age and sex, neither the fibrinogen concentration (hazard ratio [HR] =1.05, confidence interval [CI] 0.839–1.310 p=0.68), nor the –455G>A SNP (HR=0.77, 95%CI 0.491–1.197; p=0.24), nor the Val34Leu SNP (HR=0.99, 95%CI 0.646–1.542 p=0.99) were associated with occurrence of VTE. The fibrinogen concentration was not significantly different among the fibrinogen –455G or A genotype carriers (p = 0.33). Disseminated intravascular coagulation was observed in only five patients, none of these developed VTE. In conclusion, fibrinogen plasma levels, the fibrinogen –455G>A and the FXIII-A Val34Leu polymorphisms were not associated with VTE in our study.
Collapse
|
146
|
Ang L, Behnamfar O, Palakodeti S, Lin F, Pourdjabbar A, Patel MP, Reeves RR, Mahmud E. Elevated Baseline Serum Fibrinogen: Effect on 2-Year Major Adverse Cardiovascular Events Following Percutaneous Coronary Intervention. J Am Heart Assoc 2017; 6:JAHA.117.006580. [PMID: 29151032 PMCID: PMC5721757 DOI: 10.1161/jaha.117.006580] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background Elevated fibrinogen is associated with short‐term major adverse cardiovascular events (MACE) after percutaneous coronary intervention, but the relation with late MACE is unknown. Methods and Results Baseline demographics and 2‐year MACE were recorded among subjects undergoing nonemergent percutaneous coronary intervention. A total of 332 subjects (66.6±19.5 years, 69.9% male, 25.3% acute coronary syndrome) were enrolled. Two‐year MACE (periprocedural myocardial infarction 9.0%, rehospitalization 6.3%, revascularization 12.7%, non–periprocedural myocardial infarction 4.5%, stent thrombosis 0.9%, stroke 1.8%, and death 0.6%) were associated with higher fibrinogen (352.8±123.4 mg/dL versus 301.6±110.8 mg/dL; P<0.001), longer total stent length (40.1±25.3 mm versus 32.1±19.3 mm; P=0.004), acute coronary syndrome indication (38.7% versus 17.8%; P<0.001), number of bare‐metal stents (0.5±1.1 versus 0.2±0.5; P=0.002), and stent diameter ≤2.5 mm (55.8% versus 38.4%, P=0.003). No relation between platelet reactivity and 2‐year MACE was observed. Fibrinogen ≥280 mg/dL (odds ratio [OR] 3.0, confidence interval [CI], 1.6–5.4, P<0.001), total stent length ≥32 mm (OR 2.2, CI, 1.3–3.8, P<0.001), acute coronary syndrome indication (OR 4.1, CI, 2.3–7.5, P<0.001), any bare‐metal stents (OR 3.2, CI, 1.6–6.1, P<0.001), and stent diameter ≤2.5 mm (OR 2.0, CI, 1.2–3.5, P=0.010) were independently associated with 2‐year MACE. Following a landmark analysis excluding periprocedural myocardial infarction, fibrinogen ≥280 mg/dL remained strongly associated with 2‐year MACE (37.0% versus 17.4%, log‐rank P<0.001). Conclusions Elevated baseline fibrinogen level is associated with 2‐year MACE after percutaneous coronary intervention. Acute coronary syndrome indication for percutaneous coronary intervention, total stent length implanted, and use of bare‐metal stents or smaller‐diameter stents are also independently associated with 2‐year MACE, while measures of on‐thienopyridine platelet reactivity are not.
Collapse
Affiliation(s)
- Lawrence Ang
- Division of Cardiovascular Medicine, San Diego Sulpizio Cardiovascular Center, University of California, La Jolla, CA
| | - Omid Behnamfar
- Division of Cardiovascular Medicine, San Diego Sulpizio Cardiovascular Center, University of California, La Jolla, CA
| | - Samhita Palakodeti
- Division of Cardiovascular Medicine, San Diego Sulpizio Cardiovascular Center, University of California, La Jolla, CA
| | - Felice Lin
- Division of Cardiovascular Medicine, San Diego Sulpizio Cardiovascular Center, University of California, La Jolla, CA
| | - Ali Pourdjabbar
- Division of Cardiovascular Medicine, San Diego Sulpizio Cardiovascular Center, University of California, La Jolla, CA
| | - Mitul P Patel
- Division of Cardiovascular Medicine, San Diego Sulpizio Cardiovascular Center, University of California, La Jolla, CA
| | - Ryan R Reeves
- Division of Cardiovascular Medicine, San Diego Sulpizio Cardiovascular Center, University of California, La Jolla, CA
| | - Ehtisham Mahmud
- Division of Cardiovascular Medicine, San Diego Sulpizio Cardiovascular Center, University of California, La Jolla, CA
| |
Collapse
|
147
|
Cronjé HT, Nienaber-Rousseau C, Zandberg L, de Lange Z, Green FR, Pieters M. Fibrinogen and clot-related phenotypes determined by fibrinogen polymorphisms: Independent and IL-6-interactive associations. PLoS One 2017; 12:e0187712. [PMID: 29099861 PMCID: PMC5669433 DOI: 10.1371/journal.pone.0187712] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/24/2017] [Indexed: 12/04/2022] Open
Abstract
Interleukin-6 (IL-6) induces the expression of fibrinogen, and polymorphic variation within the fibrinogen genes is believed to alter the magnitude of this expression. The identification of the functional relevance of individual fibrinogen single nucleotide polymorphisms (SNPs) has been hindered by the high linkage disequilibrium (LD) reported in the European fibrinogen gene locus. This study investigated two novel and 12 known fibrinogen SNPs of potential functional relevance, in 2010 Tswana individuals known to have low LD. We aimed to identify functional polymorphisms that contribute to clot-related phenotypes and total and γ’ fibrinogen concentrations independently and through their interaction with IL-6, by taking advantage of the high fibrinogen and IL-6 concentrations and the low LD reported in black South Africans. Fibrinogen was significantly associated with IL-6, thereby mediating associations of IL-6 with clot formation and structure, although attenuating the association of IL-6 with clot lysis time. None of the common European fibrinogen haplotypes was present in this study population. Putative functional fibrinogen SNPs FGB–rs7439150, rs1800789 (–1420G/A) and rs1800787 (–148C/T) were significantly associated with fibrinogen concentration and altered clot properties, with several associations significantly influenced by IL-6 concentrations. The impact of harbouring several minor fibrinogen SNP alleles on the association of IL-6 and fibrinogen concentration was cumulative, with possession of each additional minor allele showing a stronger relationship of IL-6 with fibrinogen. This was also reflected in differences in clot properties, suggesting potential clinical relevance. Therefore, when investigating the effect of fibrinogen genetics on fibrinogen concentrations and CVD outcome, the possible interactions with modulating factors and the fact that SNP effects seem to be additive should be taken into account.
Collapse
Affiliation(s)
- H. Toinét Cronjé
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | | | - Lizelle Zandberg
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | - Zelda de Lange
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | - Fiona R. Green
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
| | - Marlien Pieters
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
- * E-mail:
| |
Collapse
|
148
|
Bekos C, Grimm C, Brodowicz T, Petru E, Hefler L, Reimer D, Koch H, Reinthaller A, Polterauer S, Polterauer M. Prognostic role of plasma fibrinogen in patients with uterine leiomyosarcoma - a multicenter study. Sci Rep 2017; 7:14474. [PMID: 29101329 PMCID: PMC5670177 DOI: 10.1038/s41598-017-13934-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 10/03/2017] [Indexed: 02/07/2023] Open
Abstract
Fibrinogen has an important pathophysiological role in tumor cell progression and development of metastases in different types of cancer. The present study aimed to evaluate the role of pre-treatment fibrinogen plasma concentrations as a biomarker for tumor biology and prognosis in patients with uterine leiomyosarcoma (ULMS). Clinical data of patients with ULMS were assessed in this multi-center study Pre-therapeutic fibrinogen plasma concentrations were evaluated. We investigated the association between fibrinogen plasma levels and clinico-pathological parameters and performed univariate and multivariable survival analyses. In total, 70 women with ULMS were included into the analysis. Mean (SD) pre-treatment fibrinogen plasma levels were 480.2 (172.3) mg/dL. Patients with advanced tumor stage, increased tumor size and higher histological grading had higher fibrinogen levels (p = 0.02, p = 0.013, and p = 0.029, respectively). In ULMS patients with increased fibrinogen levels 5-year overall survival (OS) rates were 25.0% compared to 52.9% in ULMS patients with normal fibrinogen, respectively. Univariate survival analyses revealed that elevated fibrinogen plasma levels (p = 0.030), advanced tumor stage (p < 0.001) and undifferentiated histology (p = 0.003) showed association with unfavorable OS. In multivariable analysis, histological grade (p = 0.03) and tumor stage (0.02) were independently associated with survival. Elevated fibrinogen plasma levels were associated with aggressive tumor biology and poor prognosis in women with ULMS. Fibrinogen might be useful as a novel biomarker in ULMS.
Collapse
Affiliation(s)
- Christine Bekos
- Department of General Gynaecology and Gynaecological Oncology, Gynecologic Cancer Unit - Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Christoph Grimm
- Department of General Gynaecology and Gynaecological Oncology, Gynecologic Cancer Unit - Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Thomas Brodowicz
- Clinical Division of Oncology, Department of Medicine 1, Comprehensive Cancer Center - Medical University Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Edgar Petru
- Department of Obstetrics and Gynaecology of the Medical University of Graz, Graz, Austria
| | - Lukas Hefler
- Department of Gynaecology, Barmherzige Schwestern Hospital Linz, Linz, Austria
| | - Daniel Reimer
- Department of Obstetrics and Gynaecology of the Medical University of Innsbruck, Tirol, Austria
| | - Horst Koch
- Department of Obstetrics and Gynaecology of the Medical University of Salzburg, Salzburg, Austria
| | - Alexander Reinthaller
- Department of General Gynaecology and Gynaecological Oncology, Gynecologic Cancer Unit - Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Karl Landsteiner Institute for General Gynecology and Experimental Gynecologic Oncology, Vienna, Austria
| | - Stephan Polterauer
- Department of General Gynaecology and Gynaecological Oncology, Gynecologic Cancer Unit - Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria. .,Karl Landsteiner Institute for General Gynecology and Experimental Gynecologic Oncology, Vienna, Austria.
| | - Mariella Polterauer
- Department of General Gynaecology and Gynaecological Oncology, Gynecologic Cancer Unit - Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| |
Collapse
|
149
|
Tuleta I, Skowasch D, Aurich F, Eckstein N, Schueler R, Pizarro C, Schahab N, Nickenig G, Schaefer C, Pingel S. Asthma is associated with atherosclerotic artery changes. PLoS One 2017; 12:e0186820. [PMID: 29073174 PMCID: PMC5658104 DOI: 10.1371/journal.pone.0186820] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 10/09/2017] [Indexed: 12/18/2022] Open
Abstract
Asthma is a chronic airway inflammation with a potential systemic impact. Atherosclerosis is a chronic inflammatory artery disease. The aim of our study was to prove if there is a correlation between the occurrence of asthma and increased atherosclerotic vessel disorders. Vessel status was compared between mild-to-moderate, severe allergic asthma and matched controls. Measurements of artery stiffness were calculated by central pulse wave velocity, ultrasonographic strain imaging and ankle-brachial index. Atherosclerotic plaque burden was assessed by colour-coded duplex sonography. Additionally, analysis of cardiovascular and asthma blood markers was conducted. Arterial stiffness expressed as an increased central pulse wave velocity and decreased circumferential and radial strains as well as the prevalence of media sclerosis were significantly higher among asthma patients compared to controls. Atherosclerotic plaque burden was relevantly increased in asthma groups vs. controls (severe asthma: 43.1%, mild-to-moderate asthma: 25.0%, control: 14.3% of study participants). Except for the elevated IgE and fibrinogen concentrations as well as leukocyte number there were no relevant differences in the blood parameters between the groups. Allergic asthma is associated with distinct atherosclerotic artery changes compared to the respectively matched control collective. The severity of asthma correlates with more pronounced pathological vessel alternations.
Collapse
Affiliation(s)
- Izabela Tuleta
- Department of Internal Medicine II–Cardiology, Pulmonology and Angiology, University of Bonn, Bonn, Germany
- * E-mail:
| | - Dirk Skowasch
- Department of Internal Medicine II–Cardiology, Pulmonology and Angiology, University of Bonn, Bonn, Germany
| | - Florian Aurich
- Department of Internal Medicine II–Cardiology, Pulmonology and Angiology, University of Bonn, Bonn, Germany
| | - Nicolas Eckstein
- Department of Internal Medicine II–Cardiology, Pulmonology and Angiology, University of Bonn, Bonn, Germany
| | - Robert Schueler
- Department of Internal Medicine II–Cardiology, Pulmonology and Angiology, University of Bonn, Bonn, Germany
| | - Carmen Pizarro
- Department of Internal Medicine II–Cardiology, Pulmonology and Angiology, University of Bonn, Bonn, Germany
| | - Nadjib Schahab
- Department of Internal Medicine II–Cardiology, Pulmonology and Angiology, University of Bonn, Bonn, Germany
| | - Georg Nickenig
- Department of Internal Medicine II–Cardiology, Pulmonology and Angiology, University of Bonn, Bonn, Germany
| | - Christian Schaefer
- Department of Internal Medicine II–Cardiology, Pulmonology and Angiology, University of Bonn, Bonn, Germany
| | - Simon Pingel
- Department of Internal Medicine II–Cardiology, Pulmonology and Angiology, University of Bonn, Bonn, Germany
| |
Collapse
|
150
|
Gao XY, Zhou BY, Zhang MZ, Zhao X, Qing P, Zhu CG, Wu NQ, Guo YL, Gao Y, Li XL, Wang Y, Liu G, Dong Q, Guo LH, Li JJ. Association between fibrinogen level and the severity of coronary stenosis in 418 male patients with myocardial infarction younger than 35 years old. Oncotarget 2017; 8:81361-81368. [PMID: 29113395 PMCID: PMC5655290 DOI: 10.18632/oncotarget.18578] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 05/21/2017] [Indexed: 12/21/2022] Open
Abstract
Fibrinogen (Fib) is a useful marker for predicting the severity of coronary artery disease (CAD) in adult population. However, whether Fib can be a predictor for the presence and severity of CAD in very young MI patients (≤35 years old) remains to be determined. A total of 418 males from 61,863 patients with MI who were under 35 years old were sequentially recruited in our study. The patients were divided into two main groups and three subgroups according to coronary angiograph and Gensini score (GS) system: no coronary artery stenosis (group A), the results of the coronary artery stenosis (group B); low GS, intermediate GS and high GS. Data indicated that Fib, body mass index, current smoking, white blood cell count (WBCC) and GS were significantly higher in group B than those in group A (all P < 0.01). Moreover, there were significant differences in Fib, mean age, diabetes mellitus, family history of CAD, WBCC, left ventricular ejection fraction, and GS between high GS and low GS subgroups (all P < 0.01). A positive correlation between Fib levels and GS was found (r = 0.242, p < 0.001). Receiver operating characteristics curve analysis demonstrated that the best cut-off level of Fib predicting the severity of coronary stenosis was 3.475g/L (sensitivity 64%; specificity 70%) and the area under the curve was 0.656. Fib was also independently associated with high GS (OR=2.173, 95%CI 1.011-4.670, P = 0.047) after adjusting for potential confounders. In conclusion, Fib is significantly related to the presence and severity of coronary stenosis in male patients with MI under 35 years old.
Collapse
Affiliation(s)
- Xiong-Yi Gao
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- Division of Chest Pain Center, Guangdong Provincial Hospital of Chinese Medicine, The 2nd Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Bing-Yang Zhou
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Min-Zhou Zhang
- Division of Chest Pain Center, Guangdong Provincial Hospital of Chinese Medicine, The 2nd Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Xi Zhao
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Ping Qing
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Cheng-Gang Zhu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Na-Qiong Wu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Yuan-Lin Guo
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Ying Gao
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Xiao-Lin Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Yao Wang
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Geng Liu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Qian Dong
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Li-Heng Guo
- Division of Chest Pain Center, Guangdong Provincial Hospital of Chinese Medicine, The 2nd Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Jian-Jun Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| |
Collapse
|