201
|
Pop D, Dădârlat A, Zdrenghea D. Novel cardiovascular risk markers in women with ischaemic heart disease. Cardiovasc J Afr 2015; 25:137-41. [PMID: 25000444 PMCID: PMC4120125 DOI: 10.5830/cvja-2014-014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 03/14/2014] [Indexed: 11/06/2022] Open
Abstract
Abstract The incidence of coronary heart disease in premenopausal women is lower than in men because of their hormonal protection. Angina pectoris occurs in women about 10 years later than in men. However, mortality from ischaemic heart disease remains higher in women than in men. Current studies are focusing on novel cardiovascular risk biomarkers because it seems that traditional cardiovascular risk factors and their assessment scores underestimate the risk in females. Increased plasma levels of these newly established biomarkers of risk have been found to worsen endothelial dysfunction and inflammation, both of which play a key role in the pathogenesis of microvascular angina, which is very common in women. These novel cardiovascular risk markers can be classified into three categories: inflammatory markers, markers of haemostasis, and other biomarkers.
Collapse
Affiliation(s)
- Dana Pop
- University of Medicine and Pharmacy Iuliu HaŢieganu, Cluj-Napoca, Romania.
| | - Alexandra Dădârlat
- University of Medicine and Pharmacy Iuliu HaŢieganu, Cluj-Napoca, Romania
| | - D Zdrenghea
- University of Medicine and Pharmacy Iuliu HaŢieganu, Cluj-Napoca, Romania
| |
Collapse
|
202
|
Korshøj M, Ravn MH, Holtermann A, Hansen ÅM, Krustrup P. Aerobic exercise reduces biomarkers related to cardiovascular risk among cleaners: effects of a worksite intervention RCT. Int Arch Occup Environ Health 2015; 89:239-49. [PMID: 26139093 PMCID: PMC4724374 DOI: 10.1007/s00420-015-1067-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 06/16/2015] [Indexed: 11/25/2022]
Abstract
Purpose Blue-collar workers have an increased risk of cardiovascular disease. Accordingly, elevated levels of biomarkers related to risk of cardiovascular disease, such as high-sensitive C-reactive protein, have been observed among blue-collar workers. The objective was to examine whether an aerobic exercise worksite intervention changes the level of inflammation biomarkers among cleaners. Methods The design was a cluster-randomized controlled trial with 4-month worksite intervention. Before the 116 cleaners aged 18–65 years were randomized, they signed an informed consent form. The reference group (n = 59) received lectures, and the aerobic exercise group (n = 57) performed worksite aerobic exercise (30 min twice a week). Levels of biomarkers (high-sensitive C-reactive protein, fibrinogen, cholesterol, low- and high-density lipoprotein cholesterol and triglyceride) were collected at baseline and after 4 months. A repeated-measure, multi-adjusted, mixed-model intention-to-treat analysis was applied to compare between-group differences. The study was registered as ISRCTN86682076. Results Significant (p < 0.05) between-group reductions from baseline to follow-up were found for high-sensitive C-reactive protein (−0.54 ± 0.20 µg/ml; 95 % CI −0.94, −0.14), low-density lipoprotein cholesterol (−0.32 ± 0.11 mmol/L; 95 % CI −0.54, −0.10) and the ratios of LDL/HDL (−0.30 ± 0.08; 95 % CI −0.46, −0.14), and LDL/TC cholesterol (−0.04 ± 0.02; 95 % CI −0.07, −0.01). Conclusion This study indicates that an aerobic exercise intervention among cleaners leads to reduced levels of high-sensitive C-reactive protein and low-density lipoprotein cholesterol, and an unaltered level of fibrinogen. The aerobic exercise seems to improve inflammatory levels and lipoprotein profile among cleaners, with no signs of cardiovascular overload.
Collapse
Affiliation(s)
- Mette Korshøj
- National Research Centre for the Working Environment, Lersø Parkallé 105, 2100, Copenhagen Ø, Denmark. .,Department of Nutrition, Exercise and Sports, Copenhagen Centre for Team Sport and Health, University of Copenhagen, Nørre Allé 51, Copenhagen N, 2200, Denmark.
| | - Marie Højbjerg Ravn
- National Research Centre for the Working Environment, Lersø Parkallé 105, 2100, Copenhagen Ø, Denmark
| | - Andreas Holtermann
- National Research Centre for the Working Environment, Lersø Parkallé 105, 2100, Copenhagen Ø, Denmark.,Institute of Sports Science and Clinical Biomechanics, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Åse Marie Hansen
- National Research Centre for the Working Environment, Lersø Parkallé 105, 2100, Copenhagen Ø, Denmark.,Department of Public Health, University of Copenhagen, Øster Farimagsgade 5, Copenhagen K, 1014, Denmark
| | - Peter Krustrup
- Department of Nutrition, Exercise and Sports, Copenhagen Centre for Team Sport and Health, University of Copenhagen, Nørre Allé 51, Copenhagen N, 2200, Denmark.,Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, St. Luke's Campus, Exeter, UK
| |
Collapse
|
203
|
Cho HM, Kang DR, Kim HC, Oh SM, Kim BK, Suh I. Association between Fibrinogen and Carotid Atherosclerosis According to Smoking Status in a Korean Male Population. Yonsei Med J 2015; 56:921-7. [PMID: 26069112 PMCID: PMC4479858 DOI: 10.3349/ymj.2015.56.4.921] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
PURPOSE Although inconsistent, reports have shown fibrinogen levels to be associated with atherosclerosis. Accordingly, since cigarette smoking is associated with increased levels of fibrinogen and atherosclerosis, it may also affect the association between fibrinogen and atherosclerosis. We investigated the associations between fibrinogen and carotid intima-media thickness (IMT) according to smoking status in a Korean male population. MATERIALS AND METHODS Plasma fibrinogen levels were measured in 277 men aged 40-87 years without a history of myocardial infarction or stroke. High-resolution B-mode ultrasonography was used to examine the common carotid arteries. IMT level was analyzed both as a continuous (IMT-max, maximum value; IMT-tpm, 3-point mean value) and categorical variable (higher IMT; presence of plaque). Serial linear and logistic regression models were employed to examine the association between fibrinogen and IMT according to smoking status. RESULTS Fibrinogen levels were positively associated with IMT-max (standardized β=0.25, p=0.021) and IMT-tpm (standardized β=0.21, p=0.038), even after adjusting for age, body mass index, systolic blood pressure, fasting glucose, and total cholesterol to high-density lipoprotein cholesterol ratio in current smokers (n=75). No significant association between fibrinogen and IMT, however, was noted in former smokers (n=80) or nonsmokers (n=122). Adjusted odds ratios (95% confidence interval) for having plaque per one standard deviation higher fibrinogen level were 2.06 (1.09-3.89) for current smokers, 0.68 (0.43-1.10) for former smokers, and 1.06 (0.60-1.87) for nonsmokers. CONCLUSION Our findings suggest that cigarette smoking may modify the association between fibrinogen and carotid atherosclerosis. Further studies are required to confirm this finding in different populations.
Collapse
Affiliation(s)
- Hye Min Cho
- Department of Public Health, Yonsei University Graduate School, Seoul, Korea.; Department of Family Medicine, Seoul Medical Center, Seoul, Korea
| | - Dae Ryong Kang
- Department of Medical Humanities and Social Medicines, Ajou University School of Medicine, Suwon, Korea
| | - Hyeon Chang Kim
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Korea.
| | - Sun Min Oh
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Korea.; Medical Affairs, Novartis Korea Oncology, Seoul, Korea
| | - Byeong-Keuk Kim
- Yonsei Cardiovascular Center, Yonsei University College of Medicine, Seoul, Korea
| | - Il Suh
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
204
|
Rare and low-frequency variants and their association with plasma levels of fibrinogen, FVII, FVIII, and vWF. Blood 2015; 126:e19-29. [PMID: 26105150 DOI: 10.1182/blood-2015-02-624551] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/27/2015] [Indexed: 12/21/2022] Open
Abstract
Fibrinogen, coagulation factor VII (FVII), and factor VIII (FVIII) and its carrier von Willebrand factor (vWF) play key roles in hemostasis. Previously identified common variants explain only a small fraction of the trait heritabilities, and additional variations may be explained by associations with rarer variants with larger effects. The aim of this study was to identify low-frequency (minor allele frequency [MAF] ≥0.01 and <0.05) and rare (MAF <0.01) variants that influence plasma concentrations of these 4 hemostatic factors by meta-analyzing exome chip data from up to 76,000 participants of 4 ancestries. We identified 12 novel associations of low-frequency (n = 2) and rare (n = 10) variants across the fibrinogen, FVII, FVIII, and vWF traits that were independent of previously identified associations. Novel loci were found within previously reported genes and had effect sizes much larger than and independent of previously identified common variants. In addition, associations at KCNT1, HID1, and KATNB1 identified new candidate genes related to hemostasis for follow-up replication and functional genomic analysis. Newly identified low-frequency and rare-variant associations accounted for modest amounts of trait variance and therefore are unlikely to increase predicted trait heritability but provide new information for understanding individual variation in hemostasis pathways.
Collapse
|
205
|
Swarowska M, Polczak A, Pera J, Klimkowicz-Mrowiec A, Slowik A, Dziedzic T. Hyperfibrinogenemia predicts long-term risk of death after ischemic stroke. J Thromb Thrombolysis 2015; 38:517-21. [PMID: 25106735 PMCID: PMC4182591 DOI: 10.1007/s11239-014-1122-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In stroke patients higher levels of plasma fibrinogen are associated with increased risk of unfavourable functional outcome and short-term mortality. The aim of our study was to determine the relationship between plasma fibrinogen level and long-term risk of death in ischemic stroke patients. Seven hundred thirty six patients (median age 71; 47.1 % men) admitted to the stroke unit within 24 h after stroke were included. Plasma fibrinogen level was measured on day 1 of hospitalisation. Hyperfibrinogenemia was defined as plasma fibrinogen concentration >3.5 g/L. The maximal follow-up period was 84 months. Hyperfibrinogenemia was found in 25.0 % of patients. On multivariate logistic regression analysis, after adjustment for age, stroke severity, atrial fibrillation, smoking, white blood cell count, fever, in-hospital pneumonia and hyperglycemia, hyperfibrinogenemia was associated with increased case fatality (HR 1.71, 95 % CI 1.29–2.26, P < 0.01). Hyperfibrinogenemia predicts the long-term risk of death in ischemic stroke patients.
Collapse
Affiliation(s)
- Marta Swarowska
- Department of Neurology, Jagiellonian University Medical College, ul. Botaniczna 3, 31-503, Krakow, Poland
| | | | | | | | | | | |
Collapse
|
206
|
Ploubidis GB, Silverwood RJ, DeStavola B, Grundy E. Life-Course Partnership Status and Biomarkers in Midlife: Evidence From the 1958 British Birth Cohort. Am J Public Health 2015; 105:1596-603. [PMID: 26066911 DOI: 10.2105/ajph.2015.302644] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVES We examined the association between trajectories of partnership status over the life course and objectively measured health indicators in midlife. METHODS We used data from 4 waves (1981, 1991, 2000, and 2002-2004) of the British National Child Development Study (NCDS), a prospective cohort study that includes all people born in Britain during 1 week in March 1958 (n = 18 558). RESULTS After controlling for selection attributable to early-life and early-adulthood characteristics, we found that life-course trajectories of partnership status were associated with hemostatic and inflammatory markers, the prevalence of metabolic syndrome and respiratory function in midlife. Never marrying or cohabiting was negatively associated with health in midlife for both genders, but the effect was more pronounced in men. Women who had married in their late 20s or early 30s and remained married had the best health in midlife. Men and women in cohabiting unions had midlife health outcomes similar to those in formal marriages. CONCLUSIONS Partnership status over the life course has a cumulative effect on a wide range of objectively measured health indicators in midlife.
Collapse
Affiliation(s)
- George B Ploubidis
- George B. Ploubidis is with the Centre for Longitudinal Studies, UCL - Institute of Education, University College London, London, UK. Richard J. Silverwood and Bianca DeStavola are with the Centre for Statistical Methodology, Department of Medical Statistics, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London. Emily Grundy is with the Department of Social Policy, London School of Economics and Political Science, London
| | - Richard J Silverwood
- George B. Ploubidis is with the Centre for Longitudinal Studies, UCL - Institute of Education, University College London, London, UK. Richard J. Silverwood and Bianca DeStavola are with the Centre for Statistical Methodology, Department of Medical Statistics, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London. Emily Grundy is with the Department of Social Policy, London School of Economics and Political Science, London
| | - Bianca DeStavola
- George B. Ploubidis is with the Centre for Longitudinal Studies, UCL - Institute of Education, University College London, London, UK. Richard J. Silverwood and Bianca DeStavola are with the Centre for Statistical Methodology, Department of Medical Statistics, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London. Emily Grundy is with the Department of Social Policy, London School of Economics and Political Science, London
| | - Emily Grundy
- George B. Ploubidis is with the Centre for Longitudinal Studies, UCL - Institute of Education, University College London, London, UK. Richard J. Silverwood and Bianca DeStavola are with the Centre for Statistical Methodology, Department of Medical Statistics, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London. Emily Grundy is with the Department of Social Policy, London School of Economics and Political Science, London
| |
Collapse
|
207
|
McEvoy JW, Nasir K, DeFilippis AP, Lima JAC, Bluemke DA, Hundley WG, Barr RG, Budoff MJ, Szklo M, Navas-Acien A, Polak JF, Blumenthal RS, Post WS, Blaha MJ. Relationship of cigarette smoking with inflammation and subclinical vascular disease: the Multi-Ethnic Study of Atherosclerosis. Arterioscler Thromb Vasc Biol 2015; 35:1002-10. [PMID: 25745060 DOI: 10.1161/atvbaha.114.304960] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE We sought to assess the impact of smoking status, cumulative pack-years, and time since cessation (the latter in former smokers only) on 3 important domains of cardiovascular disease: inflammation, vascular dynamics and function, and subclinical atherosclerosis. APPROACH AND RESULTS The Multi-Ethnic Study of Atherosclerosis (MESA) cohort enrolled 6814 adults without prior cardiovascular disease. Smoking variables were determined by self-report and confirmed with urinary cotinine. We examined cross-sectional associations between smoking parameters and (1) inflammatory biomarkers (high-sensitivity C-reactive protein [hsCRP], interleukin-6, and fibrinogen); (2) vascular dynamics and function (brachial flow-mediated dilation and carotid distensibility by ultrasound, as well as aortic distensibility by MRI); and (3) subclinical atherosclerosis (coronary artery calcification, carotid intima-media thickness, and ankle-brachial index). We identified 3218 never smokers, 2607 former smokers, and 971 current smokers. Mean age was 62 years and 47% were male. There was no consistent association between smoking and vascular distensibility or flow-mediated dilation outcomes. However, compared with never smokers, the adjusted association between current smoking and measures of either inflammation or subclinical atherosclerosis was consistently stronger than for former smoking (eg, odds ratio for hsCRP>2 mg/L of 1.7 [95% confidence interval, 1.5-2.1] versus 1.2 [1.1-1.4], odds ratio for coronary artery calcification>0 of 1.8 [1.5-2.1] versus 1.4 [1.2-1.6], respectively). Similar associations were seen for interleukin-6, fibrinogen, carotid intima-media thickness, and ankle-brachial index. A monotonic association was also found between higher pack-year quartiles and increasing inflammatory markers. Furthermore, current smokers with hsCRP>2 mg/L were more likely to have increased carotid intima-media thickness, abnormal ankle-brachial index, and coronary artery calcification>75th percentile for age, sex, and race (relative to smokers with hsCRP<2 mg/L, interaction P<0.05 for all 3 outcomes). In contrast, time since quitting in former smokers was independently associated with lower inflammation and atherosclerosis (eg, odds ratio for hsCRP>2 mg/L of 0.91 [0.88-0.95] and odds ratio for coronary artery calcification>0 of 0.94 [0.90-0.97] for every 5-year cessation interval). CONCLUSIONS These findings expand our understanding of the harmful effects of smoking and help explain the cardiovascular benefits of smoking cessation.
Collapse
Affiliation(s)
- John W McEvoy
- From the Ciccarone Center for the Prevention of Heart Disease (J.W.M., K.N., A.P.D., R.S.B., M.J. Blaha) and Division of Cardiology (J.A.C.L., W.S.P.), Johns Hopkins University, Baltimore, MD; Center for Wellness and Prevention, Baptist Health South Florida, Miami Beach (K.N.); Division of Cardiology, University of Louisville, Rudd Heart and Lung Center, KY (A.P.D.); Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD (D.A.B.); Division of Cardiology, Wake Forest University Health Center, Winston-Salem, NC (W.G.H.); Division of General Medicine, Division of Pulmonary, Allergy and Critical Care, Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Los Angeles Biomedical Research Institute at Harbor-UCLA, Los Angeles, CA (M.J. Budoff); Bloomberg School of Public Health, John Hopkins University, Baltimore, MD (M.S., A.N.-A., W.S.P., M.J.B.); and Department of Radiology, Tufts University School of Medicine, Boston, MA (J.F.P.)
| | - Khurram Nasir
- From the Ciccarone Center for the Prevention of Heart Disease (J.W.M., K.N., A.P.D., R.S.B., M.J. Blaha) and Division of Cardiology (J.A.C.L., W.S.P.), Johns Hopkins University, Baltimore, MD; Center for Wellness and Prevention, Baptist Health South Florida, Miami Beach (K.N.); Division of Cardiology, University of Louisville, Rudd Heart and Lung Center, KY (A.P.D.); Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD (D.A.B.); Division of Cardiology, Wake Forest University Health Center, Winston-Salem, NC (W.G.H.); Division of General Medicine, Division of Pulmonary, Allergy and Critical Care, Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Los Angeles Biomedical Research Institute at Harbor-UCLA, Los Angeles, CA (M.J. Budoff); Bloomberg School of Public Health, John Hopkins University, Baltimore, MD (M.S., A.N.-A., W.S.P., M.J.B.); and Department of Radiology, Tufts University School of Medicine, Boston, MA (J.F.P.)
| | - Andrew P DeFilippis
- From the Ciccarone Center for the Prevention of Heart Disease (J.W.M., K.N., A.P.D., R.S.B., M.J. Blaha) and Division of Cardiology (J.A.C.L., W.S.P.), Johns Hopkins University, Baltimore, MD; Center for Wellness and Prevention, Baptist Health South Florida, Miami Beach (K.N.); Division of Cardiology, University of Louisville, Rudd Heart and Lung Center, KY (A.P.D.); Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD (D.A.B.); Division of Cardiology, Wake Forest University Health Center, Winston-Salem, NC (W.G.H.); Division of General Medicine, Division of Pulmonary, Allergy and Critical Care, Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Los Angeles Biomedical Research Institute at Harbor-UCLA, Los Angeles, CA (M.J. Budoff); Bloomberg School of Public Health, John Hopkins University, Baltimore, MD (M.S., A.N.-A., W.S.P., M.J.B.); and Department of Radiology, Tufts University School of Medicine, Boston, MA (J.F.P.)
| | - Joao A C Lima
- From the Ciccarone Center for the Prevention of Heart Disease (J.W.M., K.N., A.P.D., R.S.B., M.J. Blaha) and Division of Cardiology (J.A.C.L., W.S.P.), Johns Hopkins University, Baltimore, MD; Center for Wellness and Prevention, Baptist Health South Florida, Miami Beach (K.N.); Division of Cardiology, University of Louisville, Rudd Heart and Lung Center, KY (A.P.D.); Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD (D.A.B.); Division of Cardiology, Wake Forest University Health Center, Winston-Salem, NC (W.G.H.); Division of General Medicine, Division of Pulmonary, Allergy and Critical Care, Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Los Angeles Biomedical Research Institute at Harbor-UCLA, Los Angeles, CA (M.J. Budoff); Bloomberg School of Public Health, John Hopkins University, Baltimore, MD (M.S., A.N.-A., W.S.P., M.J.B.); and Department of Radiology, Tufts University School of Medicine, Boston, MA (J.F.P.)
| | - David A Bluemke
- From the Ciccarone Center for the Prevention of Heart Disease (J.W.M., K.N., A.P.D., R.S.B., M.J. Blaha) and Division of Cardiology (J.A.C.L., W.S.P.), Johns Hopkins University, Baltimore, MD; Center for Wellness and Prevention, Baptist Health South Florida, Miami Beach (K.N.); Division of Cardiology, University of Louisville, Rudd Heart and Lung Center, KY (A.P.D.); Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD (D.A.B.); Division of Cardiology, Wake Forest University Health Center, Winston-Salem, NC (W.G.H.); Division of General Medicine, Division of Pulmonary, Allergy and Critical Care, Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Los Angeles Biomedical Research Institute at Harbor-UCLA, Los Angeles, CA (M.J. Budoff); Bloomberg School of Public Health, John Hopkins University, Baltimore, MD (M.S., A.N.-A., W.S.P., M.J.B.); and Department of Radiology, Tufts University School of Medicine, Boston, MA (J.F.P.)
| | - W Gregory Hundley
- From the Ciccarone Center for the Prevention of Heart Disease (J.W.M., K.N., A.P.D., R.S.B., M.J. Blaha) and Division of Cardiology (J.A.C.L., W.S.P.), Johns Hopkins University, Baltimore, MD; Center for Wellness and Prevention, Baptist Health South Florida, Miami Beach (K.N.); Division of Cardiology, University of Louisville, Rudd Heart and Lung Center, KY (A.P.D.); Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD (D.A.B.); Division of Cardiology, Wake Forest University Health Center, Winston-Salem, NC (W.G.H.); Division of General Medicine, Division of Pulmonary, Allergy and Critical Care, Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Los Angeles Biomedical Research Institute at Harbor-UCLA, Los Angeles, CA (M.J. Budoff); Bloomberg School of Public Health, John Hopkins University, Baltimore, MD (M.S., A.N.-A., W.S.P., M.J.B.); and Department of Radiology, Tufts University School of Medicine, Boston, MA (J.F.P.)
| | - R Graham Barr
- From the Ciccarone Center for the Prevention of Heart Disease (J.W.M., K.N., A.P.D., R.S.B., M.J. Blaha) and Division of Cardiology (J.A.C.L., W.S.P.), Johns Hopkins University, Baltimore, MD; Center for Wellness and Prevention, Baptist Health South Florida, Miami Beach (K.N.); Division of Cardiology, University of Louisville, Rudd Heart and Lung Center, KY (A.P.D.); Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD (D.A.B.); Division of Cardiology, Wake Forest University Health Center, Winston-Salem, NC (W.G.H.); Division of General Medicine, Division of Pulmonary, Allergy and Critical Care, Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Los Angeles Biomedical Research Institute at Harbor-UCLA, Los Angeles, CA (M.J. Budoff); Bloomberg School of Public Health, John Hopkins University, Baltimore, MD (M.S., A.N.-A., W.S.P., M.J.B.); and Department of Radiology, Tufts University School of Medicine, Boston, MA (J.F.P.)
| | - Matthew J Budoff
- From the Ciccarone Center for the Prevention of Heart Disease (J.W.M., K.N., A.P.D., R.S.B., M.J. Blaha) and Division of Cardiology (J.A.C.L., W.S.P.), Johns Hopkins University, Baltimore, MD; Center for Wellness and Prevention, Baptist Health South Florida, Miami Beach (K.N.); Division of Cardiology, University of Louisville, Rudd Heart and Lung Center, KY (A.P.D.); Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD (D.A.B.); Division of Cardiology, Wake Forest University Health Center, Winston-Salem, NC (W.G.H.); Division of General Medicine, Division of Pulmonary, Allergy and Critical Care, Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Los Angeles Biomedical Research Institute at Harbor-UCLA, Los Angeles, CA (M.J. Budoff); Bloomberg School of Public Health, John Hopkins University, Baltimore, MD (M.S., A.N.-A., W.S.P., M.J.B.); and Department of Radiology, Tufts University School of Medicine, Boston, MA (J.F.P.)
| | - Moyses Szklo
- From the Ciccarone Center for the Prevention of Heart Disease (J.W.M., K.N., A.P.D., R.S.B., M.J. Blaha) and Division of Cardiology (J.A.C.L., W.S.P.), Johns Hopkins University, Baltimore, MD; Center for Wellness and Prevention, Baptist Health South Florida, Miami Beach (K.N.); Division of Cardiology, University of Louisville, Rudd Heart and Lung Center, KY (A.P.D.); Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD (D.A.B.); Division of Cardiology, Wake Forest University Health Center, Winston-Salem, NC (W.G.H.); Division of General Medicine, Division of Pulmonary, Allergy and Critical Care, Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Los Angeles Biomedical Research Institute at Harbor-UCLA, Los Angeles, CA (M.J. Budoff); Bloomberg School of Public Health, John Hopkins University, Baltimore, MD (M.S., A.N.-A., W.S.P., M.J.B.); and Department of Radiology, Tufts University School of Medicine, Boston, MA (J.F.P.)
| | - Ana Navas-Acien
- From the Ciccarone Center for the Prevention of Heart Disease (J.W.M., K.N., A.P.D., R.S.B., M.J. Blaha) and Division of Cardiology (J.A.C.L., W.S.P.), Johns Hopkins University, Baltimore, MD; Center for Wellness and Prevention, Baptist Health South Florida, Miami Beach (K.N.); Division of Cardiology, University of Louisville, Rudd Heart and Lung Center, KY (A.P.D.); Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD (D.A.B.); Division of Cardiology, Wake Forest University Health Center, Winston-Salem, NC (W.G.H.); Division of General Medicine, Division of Pulmonary, Allergy and Critical Care, Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Los Angeles Biomedical Research Institute at Harbor-UCLA, Los Angeles, CA (M.J. Budoff); Bloomberg School of Public Health, John Hopkins University, Baltimore, MD (M.S., A.N.-A., W.S.P., M.J.B.); and Department of Radiology, Tufts University School of Medicine, Boston, MA (J.F.P.)
| | - Joseph F Polak
- From the Ciccarone Center for the Prevention of Heart Disease (J.W.M., K.N., A.P.D., R.S.B., M.J. Blaha) and Division of Cardiology (J.A.C.L., W.S.P.), Johns Hopkins University, Baltimore, MD; Center for Wellness and Prevention, Baptist Health South Florida, Miami Beach (K.N.); Division of Cardiology, University of Louisville, Rudd Heart and Lung Center, KY (A.P.D.); Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD (D.A.B.); Division of Cardiology, Wake Forest University Health Center, Winston-Salem, NC (W.G.H.); Division of General Medicine, Division of Pulmonary, Allergy and Critical Care, Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Los Angeles Biomedical Research Institute at Harbor-UCLA, Los Angeles, CA (M.J. Budoff); Bloomberg School of Public Health, John Hopkins University, Baltimore, MD (M.S., A.N.-A., W.S.P., M.J.B.); and Department of Radiology, Tufts University School of Medicine, Boston, MA (J.F.P.)
| | - Roger S Blumenthal
- From the Ciccarone Center for the Prevention of Heart Disease (J.W.M., K.N., A.P.D., R.S.B., M.J. Blaha) and Division of Cardiology (J.A.C.L., W.S.P.), Johns Hopkins University, Baltimore, MD; Center for Wellness and Prevention, Baptist Health South Florida, Miami Beach (K.N.); Division of Cardiology, University of Louisville, Rudd Heart and Lung Center, KY (A.P.D.); Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD (D.A.B.); Division of Cardiology, Wake Forest University Health Center, Winston-Salem, NC (W.G.H.); Division of General Medicine, Division of Pulmonary, Allergy and Critical Care, Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Los Angeles Biomedical Research Institute at Harbor-UCLA, Los Angeles, CA (M.J. Budoff); Bloomberg School of Public Health, John Hopkins University, Baltimore, MD (M.S., A.N.-A., W.S.P., M.J.B.); and Department of Radiology, Tufts University School of Medicine, Boston, MA (J.F.P.)
| | - Wendy S Post
- From the Ciccarone Center for the Prevention of Heart Disease (J.W.M., K.N., A.P.D., R.S.B., M.J. Blaha) and Division of Cardiology (J.A.C.L., W.S.P.), Johns Hopkins University, Baltimore, MD; Center for Wellness and Prevention, Baptist Health South Florida, Miami Beach (K.N.); Division of Cardiology, University of Louisville, Rudd Heart and Lung Center, KY (A.P.D.); Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD (D.A.B.); Division of Cardiology, Wake Forest University Health Center, Winston-Salem, NC (W.G.H.); Division of General Medicine, Division of Pulmonary, Allergy and Critical Care, Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Los Angeles Biomedical Research Institute at Harbor-UCLA, Los Angeles, CA (M.J. Budoff); Bloomberg School of Public Health, John Hopkins University, Baltimore, MD (M.S., A.N.-A., W.S.P., M.J.B.); and Department of Radiology, Tufts University School of Medicine, Boston, MA (J.F.P.)
| | - Michael J Blaha
- From the Ciccarone Center for the Prevention of Heart Disease (J.W.M., K.N., A.P.D., R.S.B., M.J. Blaha) and Division of Cardiology (J.A.C.L., W.S.P.), Johns Hopkins University, Baltimore, MD; Center for Wellness and Prevention, Baptist Health South Florida, Miami Beach (K.N.); Division of Cardiology, University of Louisville, Rudd Heart and Lung Center, KY (A.P.D.); Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD (D.A.B.); Division of Cardiology, Wake Forest University Health Center, Winston-Salem, NC (W.G.H.); Division of General Medicine, Division of Pulmonary, Allergy and Critical Care, Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Los Angeles Biomedical Research Institute at Harbor-UCLA, Los Angeles, CA (M.J. Budoff); Bloomberg School of Public Health, John Hopkins University, Baltimore, MD (M.S., A.N.-A., W.S.P., M.J.B.); and Department of Radiology, Tufts University School of Medicine, Boston, MA (J.F.P.).
| |
Collapse
|
208
|
Wannamethee SG, Whincup PH, Lennon L, Papacosta O, Lowe GD. Associations between fibrin D-dimer, markers of inflammation, incident self-reported mobility limitation, and all-cause mortality in older men. J Am Geriatr Soc 2015; 62:2357-62. [PMID: 25516032 PMCID: PMC4293158 DOI: 10.1111/jgs.13133] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVES To examine the independent relationships between fibrin D-dimer, interleukin 6 (IL-6), C-reactive protein (CRP), and fibrinogen and incident mobility limitation and mortality. DESIGN Prospective. SETTING General practice in 24 British towns. PARTICIPANTS Men aged 60 to 79 without prevalent heart failure followed up for an average of 11.5 years (N = 3,925). MEASUREMENTS All-cause mortality (n = 1,286) and self-reported mobility disability obtained at examination in 1998 to 2000 and in a postal questionnaire 3 to 5 years later in 2003. RESULTS High D-dimer (top vs lowest tertile: adjusted odds ratio (aOR) = 1.46, 95% confidence interval = 1.02-2.05) and IL-6 (aOR = 1.43, 95% CI = 1.01-2.02) levels (but not CRP or fibrinogen) were associated with greater incident mobility limitation after adjustment for confounders and prevalent disease status. IL-6, CRP, fibrinogen, and D-dimer were significantly associated with total mortality after adjustment for confounders. Only D-dimer and IL-6 predicted total mortality independent of each other and the other biomarkers. The adjusted hazard ratio (aHR) was 1.16 (95% CI = 1.10-1.22) for a standard deviation increase in log D-dimer and 1.10 (95% CI = 1.04-1.18) for a standard deviation increase in log IL-6. D-dimer was independently related to vascular and nonvascular mortality, and IL-6 was independently related to vascular mortality. Risks of mobility limitation and mortality were greatest in those with a combination of high D-dimer and IL-6 levels. CONCLUSION D-dimer and IL-6 are associated with risk of mobility limitation and mortality in older men without heart failure. The findings suggest that coagulation leads to functional decline and mortality s that inflammation does not explain.
Collapse
Affiliation(s)
- S Goya Wannamethee
- Department of Primary Care and Population Health, University College London, London, UK
| | | | | | | | | |
Collapse
|
209
|
Etemadifar R, Konarizadeh S, Zarei A, Farshidi H, Sobhani A. Relationship between periodontal status and C-reactive protein and interleuckin-6 levels among atherosclerotic patients in Bandar Abbas, Iran in 2014. Electron Physician 2015; 7:1010-6. [PMID: 26052413 PMCID: PMC4455295 DOI: 10.14661/2015.1010-1016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 08/26/2014] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Recent studies have reported an association between periodontitis and cardiovascular diseases. Atherosclerosis is also a risk factor for cardiovascular diseases. IL-6 and CRP are important inflammatory markers that are important because they have been shown to be higher when a patient has periodontitis, and they are related to atherosclerosis. The aim of this study was to assess the relationship between periodontitis and CRP and IL-6 in atherosclerotic patients. METHODS The study population in this case control study was atherosclerotic patients in Bandar Abbas, Iran in 2014. The participants included 30 individuals with periodontal diseases and 30 individuals without periodontal diseases, and they were allocated into two groups according to probe depth (PD) and clinical attachment loss (CAL). Inflammatory markers, including CRP and IL-6 were measured in the two groups. The data were analyzed using IBM SPSS 21 statistical software. Descriptive statistics, chi-squared, independent samples t-test, and Mann-Whitney tests were used to analyze the data. RESULTS Individuals with abnormal CRP had significantly higher PD and CAL than individuals with normal CRP (P<0.001). Although PD was not significantly different in individuals with normal and abnormal IL-6 (P=0.124), CAL was significantly higher in individuals with abnormal IL-6 than in the other individuals (P=0.005). CONCLUSION The results of this study showed that CRP and IL-6 are associated with periodontal diseases in atherosclerotic patients. Treatment of periodontal diseases is recommended in atherosclerotic patients.
Collapse
Affiliation(s)
- Ruhollah Etemadifar
- Periodontologist, Assistant Professor, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Shokufe Konarizadeh
- Dentistry Student, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Atefeh Zarei
- Dentistry Student, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Hossein Farshidi
- Cardiologist, Associate Professor, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Alireza Sobhani
- Pathologist, Assistant Professor, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
210
|
Baumert J, Huang J, McKnight B, Sabater-Lleal M, Steri M, Chu AY, Trompet S, Lopez LM, Fornage M, Teumer A, Tang W, Rudnicka AR, Mälarstig A, Hottenga JJ, Kavousi M, Lahti J, Tanaka T, Hayward C, Huffman JE, Morange PE, Rose LM, Basu S, Rumley A, Stott DJ, Buckley BM, de Craen AJM, Sanna S, Masala M, Biffar R, Homuth G, Silveira A, Sennblad B, Goel A, Watkins H, Müller-Nurasyid M, Rückerl R, Taylor K, Chen MH, de Geus EJC, Hofman A, Witteman JCM, de Maat MPM, Palotie A, Davies G, Siscovick DS, Kolcic I, Wild SH, Song J, McArdle WL, Ford I, Sattar N, Schlessinger D, Grotevendt A, Franzosi MG, Illig T, Waldenberger M, Lumley T, Tofler GH, Willemsen G, Uitterlinden AG, Rivadeneira F, Räikkönen K, Chasman DI, Folsom AR, Lowe GD, Westendorp RGJ, Slagboom PE, Cucca F, Wallaschofski H, Strawbridge RJ, Seedorf U, Koenig W, Bis JC, Mukamal KJ, van Dongen J, Widen E, Franco OH, Starr JM, Liu K, Ferrucci L, Polasek O, Wilson JF, Oudot-Mellakh T, Campbell H, Navarro P, Bandinelli S, Eriksson J, Boomsma DI, Dehghan A, Clarke R, Hamsten A, Boerwinkle E, Jukema JW, Naitza S, Ridker PM, Völzke H, Deary IJ, Reiner AP, Trégouët DA, O'Donnell CJ, et alBaumert J, Huang J, McKnight B, Sabater-Lleal M, Steri M, Chu AY, Trompet S, Lopez LM, Fornage M, Teumer A, Tang W, Rudnicka AR, Mälarstig A, Hottenga JJ, Kavousi M, Lahti J, Tanaka T, Hayward C, Huffman JE, Morange PE, Rose LM, Basu S, Rumley A, Stott DJ, Buckley BM, de Craen AJM, Sanna S, Masala M, Biffar R, Homuth G, Silveira A, Sennblad B, Goel A, Watkins H, Müller-Nurasyid M, Rückerl R, Taylor K, Chen MH, de Geus EJC, Hofman A, Witteman JCM, de Maat MPM, Palotie A, Davies G, Siscovick DS, Kolcic I, Wild SH, Song J, McArdle WL, Ford I, Sattar N, Schlessinger D, Grotevendt A, Franzosi MG, Illig T, Waldenberger M, Lumley T, Tofler GH, Willemsen G, Uitterlinden AG, Rivadeneira F, Räikkönen K, Chasman DI, Folsom AR, Lowe GD, Westendorp RGJ, Slagboom PE, Cucca F, Wallaschofski H, Strawbridge RJ, Seedorf U, Koenig W, Bis JC, Mukamal KJ, van Dongen J, Widen E, Franco OH, Starr JM, Liu K, Ferrucci L, Polasek O, Wilson JF, Oudot-Mellakh T, Campbell H, Navarro P, Bandinelli S, Eriksson J, Boomsma DI, Dehghan A, Clarke R, Hamsten A, Boerwinkle E, Jukema JW, Naitza S, Ridker PM, Völzke H, Deary IJ, Reiner AP, Trégouët DA, O'Donnell CJ, Strachan DP, Peters A, Smith NL. No evidence for genome-wide interactions on plasma fibrinogen by smoking, alcohol consumption and body mass index: results from meta-analyses of 80,607 subjects. PLoS One 2014; 9:e111156. [PMID: 25551457 PMCID: PMC4281156 DOI: 10.1371/journal.pone.0111156] [Show More Authors] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 09/23/2014] [Indexed: 11/23/2022] Open
Abstract
Plasma fibrinogen is an acute phase protein playing an important role in the blood coagulation cascade having strong associations with smoking, alcohol consumption and body mass index (BMI). Genome-wide association studies (GWAS) have identified a variety of gene regions associated with elevated plasma fibrinogen concentrations. However, little is yet known about how associations between environmental factors and fibrinogen might be modified by genetic variation. Therefore, we conducted large-scale meta-analyses of genome-wide interaction studies to identify possible interactions of genetic variants and smoking status, alcohol consumption or BMI on fibrinogen concentration. The present study included 80,607 subjects of European ancestry from 22 studies. Genome-wide interaction analyses were performed separately in each study for about 2.6 million single nucleotide polymorphisms (SNPs) across the 22 autosomal chromosomes. For each SNP and risk factor, we performed a linear regression under an additive genetic model including an interaction term between SNP and risk factor. Interaction estimates were meta-analysed using a fixed-effects model. No genome-wide significant interaction with smoking status, alcohol consumption or BMI was observed in the meta-analyses. The most suggestive interaction was found for smoking and rs10519203, located in the LOC123688 region on chromosome 15, with a p value of 6.2×10−8. This large genome-wide interaction study including 80,607 participants found no strong evidence of interaction between genetic variants and smoking status, alcohol consumption or BMI on fibrinogen concentrations. Further studies are needed to yield deeper insight in the interplay between environmental factors and gene variants on the regulation of fibrinogen concentrations.
Collapse
Affiliation(s)
- Jens Baumert
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Jie Huang
- National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, United States of America
- National Heart, Lung and Blood Institute Division of Intramural Research, Bethesda, Maryland, United States of America
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Barbara McKnight
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Maria Sabater-Lleal
- Cardiovascular Genetics and Genomics Group, Atherosclerosis Research Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Maristella Steri
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cagliari, Italy
| | - Audrey Y. Chu
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Lorna M. Lopez
- Department of Psychology, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, Division of Epidemiology, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Alexander Teumer
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Weihong Tang
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Alicja R. Rudnicka
- Division of Population Health Sciences & Education, St George's, University of London, Cranmer Terrace, London, United Kingdom
| | - Anders Mälarstig
- Cardiovascular Genetics and Genomics Group, Atherosclerosis Research Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, VU University & EMGO+ institute, VU Medical Centre, Amsterdam, the Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Netherlands Genomics Initiative (NGI)-Sponsored Netherlands Consortium for Healthy Aging (NCHA), Rotterdam, the Netherlands
| | - Jari Lahti
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland
- Folkhalsan Research Centre, Helsinki, Finland
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, Scotland, United Kingdom
| | - Jennifer E. Huffman
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, Scotland, United Kingdom
| | | | - Lynda M. Rose
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Saonli Basu
- Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ann Rumley
- Division of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - David J. Stott
- Institute of Cardiovascular and Medical Sciences, Faculty of Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Brendan M. Buckley
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Anton J. M. de Craen
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Serena Sanna
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cagliari, Italy
| | - Marco Masala
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cagliari, Italy
| | - Reiner Biffar
- Department of Prosthetic Dentistry, Gerostomatology and Dental Materials, University Medicine Greifswald, Greifswald, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Angela Silveira
- Cardiovascular Genetics and Genomics Group, Atherosclerosis Research Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Bengt Sennblad
- Cardiovascular Genetics and Genomics Group, Atherosclerosis Research Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
- Science for Life Laboratory, Karolinska Insitutet, Stockholm, Sweden
| | - Anuj Goel
- Department of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
- Department of Cardiovascular Medicine, The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Hugh Watkins
- Department of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
- Department of Cardiovascular Medicine, The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Regina Rückerl
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- ESC-Environmental Science Center, University of Augsburg, Augsburg, Germany
| | - Kent Taylor
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Ming-Huei Chen
- Department of Biostatistics, Boston University, Boston, Massachusetts, United States of America
| | - Eco J. C. de Geus
- Department of Haematology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Netherlands Genomics Initiative (NGI)-Sponsored Netherlands Consortium for Healthy Aging (NCHA), Rotterdam, the Netherlands
| | - Jacqueline C. M. Witteman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Netherlands Genomics Initiative (NGI)-Sponsored Netherlands Consortium for Healthy Aging (NCHA), Rotterdam, the Netherlands
| | | | - Aarno Palotie
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, Finland
| | - Gail Davies
- Department of Psychology, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - David S. Siscovick
- Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Ivana Kolcic
- Department of Public Health, University of Split Medical School, Split, Croatia
| | - Sarah H. Wild
- Centre for Population Health Sciences, University of Edinburgh, Teviot Place, Edinburgh, Scotland, United Kingdom
| | - Jaejoon Song
- Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Wendy L. McArdle
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Ian Ford
- Robertson Center for Biostatistics, University of Glasgow, Glasgow, United Kingdom
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, Faculty of Medicine, Glasgow, United Kingdom
| | - David Schlessinger
- Intramural Research Program, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Anne Grotevendt
- Institute for Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Maria Grazia Franzosi
- Department of Cardiovascular Research, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Lumley
- Department of Statistics, University of Auckland, Auckland, New Zealand
| | | | - Gonneke Willemsen
- Department of Biological Psychology, VU University & EMGO+ institute, VU Medical Centre, Amsterdam, the Netherlands
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Netherlands Genomics Initiative (NGI)-Sponsored Netherlands Consortium for Healthy Aging (NCHA), Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Fernando Rivadeneira
- Division of Preventive Medicine, Division of Genetics, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Katri Räikkönen
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland
| | - Daniel I. Chasman
- Division of Preventive Medicine, Division of Genetics, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Aaron R. Folsom
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Gordon D. Lowe
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Rudi G. J. Westendorp
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - P. Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cagliari, Italy
| | - Henri Wallaschofski
- Intramural Research Program, National Institute on Aging, Baltimore, Maryland, United States of America
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Rona J. Strawbridge
- Cardiovascular Genetics and Genomics Group, Atherosclerosis Research Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Udo Seedorf
- Leibniz-Institut für Arterioskleroseforschung an der Universität Münster, Münster, Germany
| | - Wolfgang Koenig
- Department of Internal Medicine II - Cardiology, University of Ulm Medical Center, Ulm, Germany
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Kenneth J. Mukamal
- Harvard Medical School, Boston, Massachusetts, United States of America
- Division of General Medicine and Primary Care, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Jenny van Dongen
- Department of Biological Psychology, VU University & EMGO+ institute, VU Medical Centre, Amsterdam, the Netherlands
| | - Elisabeth Widen
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - Oscar H. Franco
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Netherlands Genomics Initiative (NGI)-Sponsored Netherlands Consortium for Healthy Aging (NCHA), Rotterdam, the Netherlands
| | - John M. Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
- Alzheimer Scotland Dementia Research Centre, The University of Edinburgh, Edinburgh, United Kingdom
| | - Kiang Liu
- Department of Preventive Medicine, Northwestern University Medical School, Chicago, Illinois, United States of America
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Ozren Polasek
- Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - James F. Wilson
- Department of Public Health, University of Split Medical School, Split, Croatia
| | - Tiphaine Oudot-Mellakh
- INSERM, UMR_S 1166, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1166, Team Genomics & Pathophysiology of Cardiovascular Diseases, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Harry Campbell
- Department of Public Health, University of Split Medical School, Split, Croatia
| | - Pau Navarro
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, Scotland, United Kingdom
| | | | - Johan Eriksson
- Folkhalsan Research Centre, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
- Helsinki University Central Hospital, Unit of General Practice, Helsinki, Finland
| | - Dorret I. Boomsma
- Department of Biological Psychology, VU University & EMGO+ institute, VU Medical Centre, Amsterdam, the Netherlands
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Netherlands Genomics Initiative (NGI)-Sponsored Netherlands Consortium for Healthy Aging (NCHA), Rotterdam, the Netherlands
| | - Robert Clarke
- Clinical Trial Service Unit, University of Oxford, Oxford, United Kingdom
| | - Anders Hamsten
- Cardiovascular Genetics and Genomics Group, Atherosclerosis Research Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Eric Boerwinkle
- Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, United States of America
| | - J. Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Durrer Center for Cardiogenetic Research, Amsterdam, the Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, the Netherlands
| | - Silvia Naitza
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cagliari, Italy
| | - Paul M. Ridker
- Division of Preventive Medicine, Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Ian J. Deary
- Department of Psychology, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Alexander P. Reiner
- Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, Finland
| | - David-Alexandre Trégouët
- INSERM, UMR_S 1166, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1166, Team Genomics & Pathophysiology of Cardiovascular Diseases, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Christopher J. O'Donnell
- National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, United States of America
- National Heart, Lung and Blood Institute Division of Intramural Research, Bethesda, Maryland, United States of America
| | - David P. Strachan
- Division of Population Health Sciences & Education, St George's, University of London, Cranmer Terrace, London, United Kingdom
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich, Munich, Germany
- * E-mail: (A. Peters); (NLS)
| | - Nicholas L. Smith
- Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, Finland
- Group Health Research Institute, Group Health Cooperative, Seattle, Washington, United States of America
- Seattle Epidemiologic Research & Information Center, Veterans Affairs Office of Research & Development, Seattle, Washington, United States of America
- * E-mail: (A. Peters); (NLS)
| |
Collapse
|
211
|
Loeffen R, Winckers K, Ford I, Jukema JW, Robertson M, Stott DJ, Spronk HM, ten Cate H, Lowe GD. Associations Between Thrombin Generation and the Risk of Cardiovascular Disease in Elderly Patients: Results From the PROSPER Study. J Gerontol A Biol Sci Med Sci 2014; 70:982-8. [PMID: 25540034 DOI: 10.1093/gerona/glu228] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Accepted: 11/12/2014] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Hypercoagulability may be an important contributor to the pathophysiology of atherosclerosis and atherothrombosis. As thrombin fulfills a central role in coagulation and links to several cellular mechanisms involved in arterial disease, we hypothesized that thrombin generation is associated with cardiovascular events in elderly patients. METHODS We studied the relationship between plasma thrombin generation and incident coronary heart disease (CHD) and stroke in the PROspective Study of Pravastatin in the Elderly at Risk (PROSPER). From this multicenter prospective cohort, 4,932 samples of subjects (70-82 years) with pre-existing vascular disease or risk factors were available for thrombin generation measurements. RESULTS Within the 3.2 years of follow-up incident stroke and CHD was observed in 227 and 545 subjects, respectively. Baseline thrombin generation was significantly decreased in subjects with incident stroke compared with subjects without: normalized peak height 71.1±40.8% versus 82.3±44.9%, p = .0002, and normalized endogenous thrombin potential 79.1±23.3% versus 87.0±24.8%, p < .0001 (mean and SDs). Thrombin generation was independently and inversely associated with stroke risk: hazard ratio 0.71 (95%CI: 0.60-0.85), 0.68 (95%CI: 0.58-0.79), for normalized peak height and normalized endogenous thrombin potential, respectively (all p < .001). In subjects with incident CHD, thrombin generation was comparable to subjects without a coronary event. Only an increased normalized peak height was significantly associated with incident CHD (hazard ratio 1.17 [95% CI: 1.06-1.28], p = .002). CONCLUSIONS We demonstrate that a delayed and decreased thrombin generation is a strong and independent predictor for stroke in elderly people at increased risk of vascular disease. However, no convincing consistent association could be demonstrated between thrombin generation and incident CHD.
Collapse
Affiliation(s)
- Rinske Loeffen
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands.
| | - Kristien Winckers
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ian Ford
- The Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Michele Robertson
- The Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - David J Stott
- Faculty of Medicine, Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - Henri M Spronk
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Hugo ten Cate
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Gordon D Lowe
- Faculty of Medicine, Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | | |
Collapse
|
212
|
Zhang Y, Zhu CG, Guo YL, Xu RX, Li S, Dong Q, Li JJ. Higher fibrinogen level is independently linked with the presence and severity of new-onset coronary atherosclerosis among Han Chinese population. PLoS One 2014; 9:e113460. [PMID: 25426943 PMCID: PMC4245131 DOI: 10.1371/journal.pone.0113460] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 10/26/2014] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Fibrinogen is a coagulation/inflammatory biomarker strongly associated with atherogenesis. However, no data is currently available regarding the association of fibrinogen level with the presence and severity of new-onset coronary atherosclerosis assessed by Gensini score (GS), particularly in Han Chinese with a large sample size. METHODS AND RESULTS We studied 2288 consecutive, new-onset subjects undergoing coronary angiography with angina-like chest pain. Clinical and laboratory data were collected. Coronary stenotic lesions were considered to be the incidence of coronary atherosclerosis. The severity of coronary stenosis was determined by the GS system. Data indicated that patients with high GS had significantly elevated fibrinogen level (p<0.001). The prevalence and severity of coronary atherosclerosis were dramatically increased according to fibrinogen tertiles. Spearman correlation analysis revealed a positive association between fibrinogen level and GS (r = 0.138, p<0.001). Multivariate logistic regression analysis demonstrated that plasma fibrinogen level was independently associated with high GS (OR = 1.275, 95% CI 1.082-1.502, p = 0.004) after adjusting for potential confounders. Moreover, fibrinogen level was also independently related to the presence of coronary atherosclerosis (fibrinogen tertile 2: OR = 1.192, 95% CI 0.889-1.598, p = 0.241; tertile 3: OR = 2.003, 95% CI 1.383-2.903, p <0.001) and high GS (fibrinogen tertile 2: OR = 1.079, 95% CI 0.833-1.397, p = 0.565; tertile 3: OR = 1.524, 95% CI 1.155-2.011, p = 0.003) in a dose-dependent manner. Receiver-operating characteristic curve analysis showed that the best fibrinogen cut-off value for predicting the severity of coronary stenosis was 3.21 g/L. CONCLUSIONS Higher fibrinogen level is independently linked with the presence and severity of new-onset coronary atherosclerosis in Han Chinese population.
Collapse
Affiliation(s)
- Yan Zhang
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, 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, Peking Union Medical College, BeiLiShi Road 167, 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, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Rui-Xia Xu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Sha Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, 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, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, 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, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| |
Collapse
|
213
|
Willeit P, Thompson SG, Agewall S, Bergström G, Bickel H, Catapano AL, Chien KL, de Groot E, Empana JP, Etgen T, Franco OH, Iglseder B, Johnsen SH, Kavousi M, Lind L, Liu J, Mathiesen EB, Norata GD, Olsen MH, Papagianni A, Poppert H, Price JF, Sacco RL, Yanez DN, Zhao D, Schminke U, Bülbül A, Polak JF, Sitzer M, Hofman A, Grigore L, Dörr M, Su TC, Ducimetière P, Xie W, Ronkainen K, Kiechl S, Rundek T, Robertson C, Fagerberg B, Bokemark L, Steinmetz H, Ikram MA, Völzke H, Lin HJ, Plichart M, Tuomainen TP, Desvarieux M, McLachlan S, Schmidt C, Kauhanen J, Willeit J, Lorenz MW, Sander D. Inflammatory markers and extent and progression of early atherosclerosis: Meta-analysis of individual-participant-data from 20 prospective studies of the PROG-IMT collaboration. Eur J Prev Cardiol 2014; 23:194-205. [PMID: 25416041 DOI: 10.1177/2047487314560664] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 10/31/2014] [Indexed: 01/09/2023]
Abstract
BACKGROUND Large-scale epidemiological evidence on the role of inflammation in early atherosclerosis, assessed by carotid ultrasound, is lacking. We aimed to quantify cross-sectional and longitudinal associations of inflammatory markers with common-carotid-artery intima-media thickness (CCA-IMT) in the general population. METHODS Information on high-sensitivity C-reactive protein, fibrinogen, leucocyte count and CCA-IMT was available in 20 prospective cohort studies of the PROG-IMT collaboration involving 49,097 participants free of pre-existing cardiovascular disease. Estimates of associations were calculated within each study and then combined using random-effects meta-analyses. RESULTS Mean baseline CCA-IMT amounted to 0.74 mm (SD = 0.18) and mean CCA-IMT progression over a mean of 3.9 years to 0.011 mm/year (SD = 0.039). Cross-sectional analyses showed positive linear associations between inflammatory markers and baseline CCA-IMT. After adjustment for traditional cardiovascular risk factors, mean differences in baseline CCA-IMT per one-SD higher inflammatory marker were: 0.0082 mm for high-sensitivity C-reactive protein (p < 0.001); 0.0072 mm for fibrinogen (p < 0.001); and 0.0025 mm for leucocyte count (p = 0.033). 'Inflammatory load', defined as the number of elevated inflammatory markers (i.e. in upper two quintiles), showed a positive linear association with baseline CCA-IMT (p < 0.001). Longitudinal associations of baseline inflammatory markers and changes therein with CCA-IMT progression were null or at most weak. Participants with the highest 'inflammatory load' had a greater CCA-IMT progression (p = 0.015). CONCLUSION Inflammation was independently associated with CCA-IMT cross-sectionally. The lack of clear associations with CCA-IMT progression may be explained by imprecision in its assessment within a limited time period. Our findings for 'inflammatory load' suggest important combined effects of the three inflammatory markers on early atherosclerosis.
Collapse
Affiliation(s)
- Peter Willeit
- The Department of Public Health and Primary Care, University of Cambridge, UK Department of Neurology, Medical University Innsbruck, Austria
| | - Simon G Thompson
- The Department of Public Health and Primary Care, University of Cambridge, UK
| | - Stefan Agewall
- Institute of Clinical Sciences, University of Oslo, and the Department of Cardiology, Oslo University Hospital Ullevål, Norway
| | - Göran Bergström
- Wallenberg Laboratory for Cardiovascular Research, University of Gothenburg, Sweden
| | - Horst Bickel
- Department of Psychiatry and Psychotherapy, University Hospital of the Technische Universität München, Germany
| | - Alberico L Catapano
- Department of Pharmacological Sciences, University of Milan, and IRCSS Multimedica Sesto S Giovanni, Milan, Italy
| | - Kuo-Liong Chien
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan Department of Internal Medicine, National Taiwan University, Taipei, Taiwan
| | - Eric de Groot
- Academic Medical Centre, Cardiology and Thoracic Surgery, and Imagelabonline and Cardiovascular, Amsterdam, The Netherlands
| | | | - Thorleif Etgen
- Department of Neurology, Kliniken Südostbayern, Klinikum Traunstein, Germany
| | - Oscar H Franco
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Bernhard Iglseder
- Department of Geriatric Medicine, Paracelsus Medical University, and the Gemeinnützige Salzburger Landeskliniken Betriebsgesellschaft GmbH, Christian-Doppler-Klinik, Salzburg, Austria
| | - Stein H Johnsen
- Department of Neurology and Neurophysiology, University Hospital of Northern Norway, and the Department of Clinical Medicine, University of Tromsø, Tromsø, Norway
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Lars Lind
- Department of Medicine, Uppsala University, Sweden
| | - Jing Liu
- Department of Epidemiology, Beijing Institute of Heart, Lung and Blood Vessel Diseases, PR China
| | - Ellisiv B Mathiesen
- Department of Neurology and Neurophysiology, University Hospital of Northern Norway, and the Department of Clinical Medicine, University of Tromsø, Tromsø, Norway
| | - Giuseppe D Norata
- Department of Pharmacological Sciences, University of Milan, and the SISA Centre for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy
| | - Michael H Olsen
- Department of Endocrinology, Centre for Individualized Medicine in Arterial Diseases, Odense University Hospital, Denmark
| | - Aikaterini Papagianni
- Department of Nephrology, Aristotle University of Thessaloniki, Hippokration General Hospital, Greece
| | - Holger Poppert
- Department of Neurology, University Hospital of the Technische Universität München, Germany
| | - Jackie F Price
- Centre for Population Health Sciences, University of Edinburgh, UK
| | - Ralph L Sacco
- Department of Neurology, Miller School of Medicine, University of Miami, FL, USA
| | - David N Yanez
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Dong Zhao
- Department of Epidemiology, Beijing Institute of Heart, Lung and Blood Vessel Diseases, PR China
| | - Ulf Schminke
- Department of Neurology, Greifswald University Clinic, Germany
| | - Alpaslan Bülbül
- Department of Neurology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Joseph F Polak
- Tufts University School of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Matthias Sitzer
- Department of Neurology, University Hospital Frankfurt, Frankfurt am Main, Germany Department of Neurology, Klinikum Herford, Germany
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| | - Liliana Grigore
- Department of Pharmacological Sciences, University of Milan, and the SISA Centre for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy
| | - Marcus Dörr
- Department B for Internal Medicine, University Medicine Greifswald, and the German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Germany
| | - Ta-Chen Su
- Department of Internal Medicine, National Taiwan University, Taipei, Taiwan
| | | | - Wuxiang Xie
- Department of Epidemiology, Beijing Institute of Heart, Lung and Blood Vessel Diseases, PR China
| | - Kimmo Ronkainen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Stefan Kiechl
- Department of Neurology, Medical University Innsbruck, Austria
| | - Tatjana Rundek
- Department of Neurology, Miller School of Medicine, University of Miami, FL, USA
| | | | - Björn Fagerberg
- Wallenberg Laboratory for Cardiovascular Research, University of Gothenburg, Sweden
| | - Lena Bokemark
- Wallenberg Laboratory for Cardiovascular Research, University of Gothenburg, Sweden
| | - Helmuth Steinmetz
- Department of Neurology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Henry Völzke
- Institute for Community Medicine, SHIP/Clinical-Epidemiological Research, Greifswald, Germany
| | - Hung-Ju Lin
- Department of Internal Medicine, National Taiwan University, Taipei, Taiwan Health Management Centre, National Taiwan University Hospital, Taipei, Taiwan
| | - Matthieu Plichart
- INSERM, U970, Université Paris Descartes, France Gerontology Department, Broca Hospital, Paris, France
| | - Tomi-Pekka Tuomainen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Moise Desvarieux
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, USA, and the École des Hautes Études en Santé Publique, and INSERM U738, Paris, France
| | - Stela McLachlan
- Centre for Population Health Sciences, University of Edinburgh, UK
| | - Caroline Schmidt
- Wallenberg Laboratory for Cardiovascular Research, University of Gothenburg, Sweden
| | - Jussi Kauhanen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Johann Willeit
- Department of Neurology, Medical University Innsbruck, Austria
| | - Matthias W Lorenz
- Department of Neurology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Dirk Sander
- Department of Neurology, Benedictus Krankenhaus Tutzing and Feldafing, Tutzing, Germany and Technische Universität München, Germany
| | | |
Collapse
|
214
|
Park KH, Han SJ, Kim HS, Kim MK, Jo SH, Kim SA, Park WJ. Impact of Framingham risk score, flow-mediated dilation, pulse wave velocity, and biomarkers for cardiovascular events in stable angina. J Korean Med Sci 2014; 29:1391-7. [PMID: 25368493 PMCID: PMC4214940 DOI: 10.3346/jkms.2014.29.10.1391] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/26/2014] [Indexed: 12/22/2022] Open
Abstract
Although the age-adjusted Framingham risk score (AFRS), flow-mediated dilation (FMD), brachial-ankle pulse wave velocity (baPWV), high-sensitivity C-reactive protein (hsCRP), fibrinogen, homocysteine, and free fatty acid (FFA) can predict future cardiovascular events (CVEs), a comparison of these risk assessments for patients with stable angina has not been reported. We enrolled 203 patients with stable angina who had been scheduled for coronary angiography (CAG). After CAG, 134 patients showed significant coronary artery disease. During 4.2 yr follow-up, 36 patients (18%) showed CVEs, including myocardial infarction, de-novo coronary artery revascularization, in-stent restenosis, stroke, and cardiovascular death. ROC analysis showed that AFRS, FMD, baPWV, and hsCRP could predict CVEs (with AUC values of 0.752, 0.707, 0.659, and 0.702, respectively, all P<0.001 except baPWV P=0.003). A Cox proportional hazard analysis showed that AFRS and FMD were independent predictors of CVEs (HR, 2.945; 95% CI, 1.572-5.522; P=0.001 and HR, 0.914; 95% CI, 0.826-0.989; P=0.008, respectively). However, there was no difference in predictive power between combining AFRS plus FMD and AFRS alone (AUC 0.752 vs. 0.763; z=1.358, P=0.175). In patients with stable angina, AFRS and FMD are independent predictors of CVEs. However, there is no additive value of FMD on the AFRS in predicting CVEs.
Collapse
Affiliation(s)
- Kyoung-Ha Park
- Cardiovascular Division, Department of Internal Medicine, Hallym University Medical Center, Anyang, Korea
| | - Sang Jin Han
- Cardiovascular Division, Department of Internal Medicine, Hallym University Medical Center, Anyang, Korea
| | - Hyun-Sook Kim
- Cardiovascular Division, Department of Internal Medicine, Hallym University Medical Center, Anyang, Korea
| | - Min-Kyu Kim
- Cardiovascular Division, Department of Internal Medicine, Hallym University Medical Center, Anyang, Korea
| | - Sang Ho Jo
- Cardiovascular Division, Department of Internal Medicine, Hallym University Medical Center, Anyang, Korea
| | - Sung-Ai Kim
- Cardiovascular Division, Department of Internal Medicine, Hallym University Medical Center, Anyang, Korea
| | - Woo Jung Park
- Cardiovascular Division, Department of Internal Medicine, Hallym University Medical Center, Anyang, Korea
| |
Collapse
|
215
|
Segal HC, Burgess AI, Poole DL, Mehta Z, Silver LE, Rothwell PM. Population-based study of blood biomarkers in prediction of subacute recurrent stroke. Stroke 2014; 45:2912-7. [PMID: 25158774 PMCID: PMC5380212 DOI: 10.1161/strokeaha.114.005592] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 07/24/2014] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND PURPOSE Risk of recurrent stroke is high in the first few weeks after transient ischemic attack or stroke and clinical risk prediction tools have only limited accuracy, particularly after the hyperacute phase. Previous studies of the predictive value of biomarkers have been small, been done in selected populations, and have not concentrated on the acute phase or on intensively treated populations. We aimed to determine the predictive value of a panel of blood biomarkers in intensively treated patients early after transient ischemic attack and stroke. METHODS We studied 14 blood biomarkers related to inflammation, thrombosis, atherogenesis, and cardiac or neuronal cell damage in early transient ischemic attack or ischemic stroke in a population-based study (Oxford Vascular Study). Biomarker levels were related to 90-day risk of recurrent stroke as hazard ratio (95% confidence interval) per decile increase, adjusted for age and sex. RESULTS Among 1292 eligible patients, there were 53 recurrent ischemic strokes within 90 days. There were moderate correlations (r=0.40-0.61; P<0.0001) between the inflammatory biomarkers and between the cell damage and thrombotic subsets. Associations with risk of early recurrent stroke were weak, with significant associations limited to interleukin-6 (adjusted hazard ratio, 1.12; 1.01-1.24; P=0.033) and C-reactive protein (adjusted hazard ratio, 1.15; 1.02-1.30; P=0.022) after adjusting for age, sex, hypertension, smoking, and diabetes mellitus although P-selectin seemed to predict stroke after transient ischemic attack (adjusted hazard ratio, 1.28; 1.00-1.63; P=0.046). CONCLUSIONS In the largest study to date, we found limited predictive use for early recurrent stroke for a panel of inflammatory, thrombotic, and cell damage biomarkers.
Collapse
Affiliation(s)
- Helen C Segal
- From the Stroke Prevention Research Unit, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom
| | - Annette I Burgess
- From the Stroke Prevention Research Unit, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom
| | - Debbie L Poole
- From the Stroke Prevention Research Unit, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom
| | - Ziyah Mehta
- From the Stroke Prevention Research Unit, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom
| | - Louise E Silver
- From the Stroke Prevention Research Unit, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom
| | - Peter M Rothwell
- From the Stroke Prevention Research Unit, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom.
| |
Collapse
|
216
|
Stack AG, Donigiewicz U, Abdalla AA, Weiland A, Casserly LF, Cronin CJ, Nguyen HT, Hannigan A. Plasma fibrinogen associates independently with total and cardiovascular mortality among subjects with normal and reduced kidney function in the general population. QJM 2014; 107:701-13. [PMID: 24633257 DOI: 10.1093/qjmed/hcu057] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The contribution of novel risk factors to mortality in chronic kidney disease remains controversial. AIM To explore the association of plasma fibrinogen with mortality among individuals with normal and reduced kidney function. METHODS We identified 9184 subjects, age 40 and over from the Third National Health and Nutrition Examination Survey (1988-94) with vital status assessed through 2006. Plasma fibrinogen was modeled as continuous variable and in quartile groups (0 to <7.7, 7.7 to <9.0, 9.0 to <10.5 and ≥ 10.5 µmol/l) with total and cardiovascular mortality across categories of glomerular filtration rate (eGFR); <60, 60-90, >90 ml/min/1.73 m(2) using Cox regression. RESULTS In multivariate analysis, the adjusted hazard ratio (HR) per 1 µmol/l (34 mg/dl) increase in fibrinogen was 1.07 [95% confidence interval (CI) 1.04-1.09] for total mortality and 1.06 (95% CI 1.03-1.09) for cardiovascular mortality. The adjusted HR for total mortality was 1.05 (1.01-1.09) for subjects with eGFR 60-90 ml/min/1.73 m(2) and 1.06 (1.02-1.10) for subjects with eGFR <60 ml/min/1.73 m(2). Subjects in the highest quartiles within each eGFR category; >90, 60-90 and <60 ml/min/1.73 m(2) experienced HRs of 1.45 (95% CI 1.03-2.03), 1.35 (95% CI 1.00-1.83) and 1.72 (95% CI 1.14-2.58), respectively, compared with subjects in the lowest quartile group. The patterns were similar for cardiovascular mortality. CONCLUSIONS Plasma fibrinogen associates with mortality among subjects with mild to moderate kidney impairment as it does in subjects with normal kidney function and should be considered a therapeutic target for cardiovascular risk reduction.
Collapse
Affiliation(s)
- A G Stack
- From the Division of Nephrology, Department of Medicine, University Hospital Limerick, Limerick, Graduate Entry Medical School, University of Limerick, Limerick and Department of Medicine, School of Medicine, National University of Ireland, Galway, Ireland From the Division of Nephrology, Department of Medicine, University Hospital Limerick, Limerick, Graduate Entry Medical School, University of Limerick, Limerick and Department of Medicine, School of Medicine, National University of Ireland, Galway, Ireland
| | - U Donigiewicz
- From the Division of Nephrology, Department of Medicine, University Hospital Limerick, Limerick, Graduate Entry Medical School, University of Limerick, Limerick and Department of Medicine, School of Medicine, National University of Ireland, Galway, Ireland
| | - A A Abdalla
- From the Division of Nephrology, Department of Medicine, University Hospital Limerick, Limerick, Graduate Entry Medical School, University of Limerick, Limerick and Department of Medicine, School of Medicine, National University of Ireland, Galway, Ireland From the Division of Nephrology, Department of Medicine, University Hospital Limerick, Limerick, Graduate Entry Medical School, University of Limerick, Limerick and Department of Medicine, School of Medicine, National University of Ireland, Galway, Ireland
| | - A Weiland
- From the Division of Nephrology, Department of Medicine, University Hospital Limerick, Limerick, Graduate Entry Medical School, University of Limerick, Limerick and Department of Medicine, School of Medicine, National University of Ireland, Galway, Ireland
| | - L F Casserly
- From the Division of Nephrology, Department of Medicine, University Hospital Limerick, Limerick, Graduate Entry Medical School, University of Limerick, Limerick and Department of Medicine, School of Medicine, National University of Ireland, Galway, Ireland From the Division of Nephrology, Department of Medicine, University Hospital Limerick, Limerick, Graduate Entry Medical School, University of Limerick, Limerick and Department of Medicine, School of Medicine, National University of Ireland, Galway, Ireland
| | - C J Cronin
- From the Division of Nephrology, Department of Medicine, University Hospital Limerick, Limerick, Graduate Entry Medical School, University of Limerick, Limerick and Department of Medicine, School of Medicine, National University of Ireland, Galway, Ireland From the Division of Nephrology, Department of Medicine, University Hospital Limerick, Limerick, Graduate Entry Medical School, University of Limerick, Limerick and Department of Medicine, School of Medicine, National University of Ireland, Galway, Ireland
| | - H T Nguyen
- From the Division of Nephrology, Department of Medicine, University Hospital Limerick, Limerick, Graduate Entry Medical School, University of Limerick, Limerick and Department of Medicine, School of Medicine, National University of Ireland, Galway, Ireland
| | - A Hannigan
- From the Division of Nephrology, Department of Medicine, University Hospital Limerick, Limerick, Graduate Entry Medical School, University of Limerick, Limerick and Department of Medicine, School of Medicine, National University of Ireland, Galway, Ireland
| |
Collapse
|
217
|
Eckel RH, Cornier MA. Update on the NCEP ATP-III emerging cardiometabolic risk factors. BMC Med 2014; 12:115. [PMID: 25154373 PMCID: PMC4283079 DOI: 10.1186/1741-7015-12-115] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 05/20/2014] [Indexed: 12/31/2022] Open
Abstract
The intent of this review is to update the science of emerging cardiometabolic risk factors that were listed in the National Cholesterol Education Program (NCEP) Adult Treatment Panel-III (ATP-III) report of 2001 (updated in 2004). At the time these guidelines were published, the evidence was felt to be insufficient to recommend these risk factors for routine screening of cardiovascular disease risk. However, the panel felt that prudent use of these biomarkers for patients at intermediate risk of a major cardiovascular event over the subsequent 10 years might help identify patients who needed more aggressive low density lipoprotein (LDL) or non-high density lipoprotein (HDL) cholesterol lowering therapy. While a number of other emerging risk factors have been identified, this review will be limited to assessing the data and recommendations for the use of apolipoprotein B, lipoprotein (a), homocysteine, pro-thrombotic factors, inflammatory factors, impaired glucose metabolism, and measures of subclinical atherosclerotic cardiovascular disease for further cardiovascular disease risk stratification.
Collapse
Affiliation(s)
- Robert H Eckel
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Anschutz Medical Campus, Mail Stop 8106, 12801 E 17th Ave, Aurora, CO 80045 USA
| | - Marc-Andre Cornier
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Anschutz Medical Campus, Mail Stop 8106, 12801 E 17th Ave, Aurora, CO 80045 USA
| |
Collapse
|
218
|
Stolz D, Meyer A, Rakic J, Boeck L, Scherr A, Tamm M. Mortality risk prediction in COPD by a prognostic biomarker panel. Eur Respir J 2014; 44:1557-70. [DOI: 10.1183/09031936.00043814] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a complex disease with various phenotypes. The simultaneous determination of multiple biomarkers reflecting different pathobiological pathways could be useful in identifying individuals with an increased risk of death.We derived and validated a combination of three biomarkers (adrenomedullin, arginine vasopressin and atrial natriuretic peptide), assessed in plasma samples of 385 patients, to estimate mortality risk in stable COPD. Biomarkers were analysed in combination and defined as high or low.In the derivation cohort (n = 142), there were 73 deaths during the 5-year follow-up. Crude hazard ratios for mortality were 3.0 (95% CI 1.8–5.1) for one high biomarker, 4.8 (95% CI 2.4–9.5) for two biomarkers and 9.6 (95% CI 3.3–28.3) for three high biomarkers compared with no elevated biomarkers. In the validation cohort (n = 243), 87 individuals died. Corresponding hazard ratios were 1.9 (95% CI 1.1–3.3), 3.1 (95% CI 1.8–5.4) and 5.4 (95% CI 2.5–11.4). Multivariable adjustment for clinical variables as well as the BODE (body mass index, airflow obstruction, dyspnoea, exercise capacity) index and stratification by the Global Initiative for Chronic Obstructive Lung Disease stages provided consistent results. The addition of the panel of three biomarkers to the BODE index generated a net reclassification improvement of 57.9% (95% CI 21.7–92.4%) and 45.9% (95% CI 13.9–75.7%) at 3 and 5 years, respectively.Simultaneously elevated levels of adrenomedullin, arginine vasopressin and atrial natriuretic peptide are associated with increased risk of death in patients with stable COPD.
Collapse
|
219
|
Martiskainen M, Oksala N, Pohjasvaara T, Kaste M, Oksala A, Karhunen PJ, Erkinjuntti T. Βeta-fibrinogen gene promoter A -455 allele associated with poor longterm survival among 55-71 years old Caucasian women in Finnish stroke cohort. BMC Neurol 2014; 14:137. [PMID: 24957141 PMCID: PMC4131463 DOI: 10.1186/1471-2377-14-137] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 06/17/2014] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Women die of stroke more often than men. After menopause, the incidence of ischemic stroke increases rapidly. Elevated fibrinogen levels and smoking have been associated with an increased risk of stroke. In gene-cluster haplotype analyses, the beta-fibrinogen (FGB) promoter -455 G/A polymorphic locus was most strongly associated with elevated plasma fibrinogen levels. We investigated whether the FGB -455 G/A polymorphism and smoking might interact with sex on longterm survival of acute stroke sufferers. METHODS The Stroke Aging Memory (SAM) cohort comprising 486 consecutive stroke patients (55-85 years, 246 men, 240 women) subjected to clinical and MRI examination was followed over 12.5 years. During this period 347 (71.4%) patients died. The genotypes of the FGB -455 G/A polymorphism were determined by PCR. RESULTS The FGB -455 G/A polymorphism genotype distributions were 64.7%, 32.1%, and 3.2% for GG, GA, and AA, respectively. During the follow-up, the FGB -455 A + genotype did not associate with survival, nor was there any genotype-by-smoking interaction on poor outcome in the total study population. However, women aged 55-71 years who carried the FGB -455 A-allele showed worse survival regardless of smoking status compared to non-smoking FGB -455 GG homozygotes (non-smokers, crude HR = 5.21, 95% CI: 1.38-19.7; smokers, crude HR = 7.03, 95% CI: 1.81-27.3). This association persisted in adjusted analyses. No such association was observed for women in the oldest age-group, nor among men. CONCLUSION The A + genotype of the FGB -455 G/A polymorphism associated with poor survival among 55-71 years old Caucasian women in the Finnish stroke cohort.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Timo Erkinjuntti
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland.
| |
Collapse
|
220
|
Choy E, Ganeshalingam K, Semb AG, Szekanecz Z, Nurmohamed M. Cardiovascular risk in rheumatoid arthritis: recent advances in the understanding of the pivotal role of inflammation, risk predictors and the impact of treatment. Rheumatology (Oxford) 2014; 53:2143-54. [PMID: 24907149 PMCID: PMC4241890 DOI: 10.1093/rheumatology/keu224] [Citation(s) in RCA: 206] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Risk of cardiovascular (CV) disease is increased among RA patients. High inflammatory burden associated with RA appears to be a key driver of the increased cardiovascular risk. Inflammation is linked with accelerated atherosclerosis and associated with a paradoxical inversion of the relationship between CV risk and lipid levels in patients with untreated RA, recently coined the lipid paradox. Furthermore, the inflammatory burden is also associated with qualitative as well as quantitative changes in lipoproteins, with the anti-inflammatory and atheroprotective roles associated with high-density lipoprotein cholesterol significantly altered. RA therapies can increase lipid levels, which may reflect the normalization of lipids due to their inflammatory-dampening effects. However, these confounding influences of inflammation and RA therapies on lipid profiles pose challenges for assessing CV risk in RA patients and interpretation of traditional CV risk scores. In this review we examine the relationship between the increased inflammatory burden in RA and CV risk, exploring how inflammation influences lipid profiles, the impact of RA therapies and strategies for identifying and monitoring CV risk in RA patients aimed at improving CV outcomes.
Collapse
Affiliation(s)
- Ernest Choy
- Section of Rheumatology, Cardiff University School of Medicine, Cardiff, UK, Global Medical Affairs, F. Hoffmann-La Roche, Basel, Switzerland, Department of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway, Department of Rheumatology, Institute of Medicine, University of Debrecen, Debrecen, Hungary and Departments of Internal Medicine and Rheumatology, VU University Medical Center, Amsterdam, The Netherlands
| | - Kandeepan Ganeshalingam
- Section of Rheumatology, Cardiff University School of Medicine, Cardiff, UK, Global Medical Affairs, F. Hoffmann-La Roche, Basel, Switzerland, Department of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway, Department of Rheumatology, Institute of Medicine, University of Debrecen, Debrecen, Hungary and Departments of Internal Medicine and Rheumatology, VU University Medical Center, Amsterdam, The Netherlands
| | - Anne Grete Semb
- Section of Rheumatology, Cardiff University School of Medicine, Cardiff, UK, Global Medical Affairs, F. Hoffmann-La Roche, Basel, Switzerland, Department of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway, Department of Rheumatology, Institute of Medicine, University of Debrecen, Debrecen, Hungary and Departments of Internal Medicine and Rheumatology, VU University Medical Center, Amsterdam, The Netherlands
| | - Zoltán Szekanecz
- Section of Rheumatology, Cardiff University School of Medicine, Cardiff, UK, Global Medical Affairs, F. Hoffmann-La Roche, Basel, Switzerland, Department of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway, Department of Rheumatology, Institute of Medicine, University of Debrecen, Debrecen, Hungary and Departments of Internal Medicine and Rheumatology, VU University Medical Center, Amsterdam, The Netherlands
| | - Michael Nurmohamed
- Section of Rheumatology, Cardiff University School of Medicine, Cardiff, UK, Global Medical Affairs, F. Hoffmann-La Roche, Basel, Switzerland, Department of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway, Department of Rheumatology, Institute of Medicine, University of Debrecen, Debrecen, Hungary and Departments of Internal Medicine and Rheumatology, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
221
|
Ribeiro DGL, Silva RP, Barboza DRMM, Lima-Júnior RCP, Ribeiro RA. Clinical correlation between N-terminal pro-B-type natriuretic peptide and angiographic coronary atherosclerosis. Clinics (Sao Paulo) 2014; 69:405-12. [PMID: 24964305 PMCID: PMC4050322 DOI: 10.6061/clinics/2014(06)07] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 10/09/2013] [Accepted: 09/14/2013] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES This study aimed to investigate the clinical correlation between angiographic coronary atherosclerosis and N-terminal pro-B-type natriuretic peptide along with other known correlated factors. METHODS In total, 153 patients with a diagnostic hypothesis of stable angina, unstable angina or acute myocardial infarction were classified as group A (patients with angiographically normal coronary arteries) or group B (patients with angiographic coronary atherosclerosis). The two groups were analyzed with respect to the following factors: gender, age, body mass index, abdominal circumference, smoking, diabetes mellitus, arterial hypertension, early family history of atherosclerosis, statin use, the presence of metabolic syndrome, clinical presentation and biochemical factors, including cholesterol, creatinine and fibrinogen plasma concentrations, monocyte counts and N-terminal pro-B-type natriuretic peptide. RESULTS Univariate analyses comparing the two groups revealed that group B patients more frequently had diabetes, used statins and had systolic dysfunction, N-terminal pro-B-type natriuretic peptide levels ≥ 250 pg/mL, fibrinogen levels >500 mg/dL and ≥ 501 monocytes/mm3 compared with group A patients (p<0.05). Nevertheless, multivariate logistic regression analysis demonstrated that the independent predictors of angiographic coronary atherosclerosis were an N-terminal pro-B-type natriuretic peptide level ≥ 250 pg/mL, diabetes mellitus and increased monocyte numbers and fibrinogen plasma concentration, regardless of the creatinine level or the presence of systolic dysfunction. CONCLUSIONS An N-terminal pro-B-type natriuretic peptide plasma concentration of ≥ 250 pg/mL is an independent predictor of angiographic coronary atherosclerosis.
Collapse
Affiliation(s)
- Demóstenes G L Ribeiro
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Ricardo P Silva
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Daniella R M M Barboza
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Roberto C P Lima-Júnior
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Ronaldo A Ribeiro
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
222
|
Muradashvili N, Benton RL, Tyagi R, Tyagi SC, Lominadze D. Elevated level of fibrinogen increases caveolae formation; role of matrix metalloproteinase-9. Cell Biochem Biophys 2014; 69:283-94. [PMID: 24307281 PMCID: PMC4020992 DOI: 10.1007/s12013-013-9797-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The role of the inflammatory agent fibrinogen (Fg) in increased pial venular permeability has been shown previously. It was suggested that an activation of matrix metalloproteinase-9 (MMP-9) is involved in Fg-induced enhanced transcytosis through endothelial cells (ECs). However, direct link between Fg, caveolae formation, and MMP-9 activity has never been shown. We hypothesized that at an elevated level, Fg enhances formation of functional caveolae through activation of MMP-9. Male wild-type (WT, C57BL/6J) or MMP-9 gene knockout (MMP9-/-) mice were infused with Fg (4 mg/ml, final blood concentration) or equal volume of phosphate buffered saline (PBS). After 2 h, mice were sacrificed and brains were collected for immunohistochemical analyses. Mouse brain ECs were treated with 4 mg/ml of Fg or PBS in the presence or absence of MMP-9 activity inhibitor, tissue inhibitor of metalloproteinases-4 (TIMP-4, 12 ng/ml). Formation of functional caveolae was assessed by confocal microscopy. Fg-induced increased formation of caveolae, which was defined by an increased co-localization of caveolin-1 (Cav-1) and plasmalemmal vesicle-associated protein-1 and was associated with an increased phosphorylation of Cav-1, was ameliorated in the presence of TIMP-4. These results suggest that at high levels, Fg enhances formation of functional caveolae that may involve Cav-1 signaling and MMP-9 activation.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY
| | - Richard L. Benton
- Department of Anatomical Sciences and Neurobiology and Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY
| | - Reeta Tyagi
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY
| | - Suresh C. Tyagi
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY
| | - David Lominadze
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY
| |
Collapse
|
223
|
Risse F, Frank RD, Weinberger AW. Thrombophilia in Patients with Retinal Vein Occlusion: A Retrospective Analysis. Ophthalmologica 2014; 232:46-52. [DOI: 10.1159/000360013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 01/27/2014] [Indexed: 11/19/2022]
|
224
|
Muradashvili N, Khundmiri SJ, Tyagi R, Gartung A, Dean WL, Lee MJ, Lominadze D. Sphingolipids affect fibrinogen-induced caveolar transcytosis and cerebrovascular permeability. Am J Physiol Cell Physiol 2014; 307:C169-79. [PMID: 24829496 DOI: 10.1152/ajpcell.00305.2013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Inflammation-induced vascular endothelial dysfunction can allow plasma proteins to cross the vascular wall, causing edema. Proteins may traverse the vascular wall through two main pathways, the paracellular and transcellular transport pathways. Paracellular transport involves changes in endothelial cell junction proteins, while transcellular transport involves caveolar transcytosis. Since both processes are associated with filamentous actin formation, the two pathways are interconnected. Therefore, it is difficult to differentiate the prevailing role of one or the other pathway during various pathologies causing an increase in vascular permeability. Using a newly developed dual-tracer probing method, we differentiated transcellular from paracellular transport during hyperfibrinogenemia (HFg), an increase in fibrinogen (Fg) content. Roles of cholesterol and sphingolipids in formation of functional caveolae were assessed using a cholesterol chelator, methyl-β-cyclodextrin, and the de novo sphingolipid synthesis inhibitor myriocin. Fg-induced formation of functional caveolae was defined by association and colocalization of Na+-K+-ATPase and plasmalemmal vesicle-associated protein-1 with use of Förster resonance energy transfer and total internal reflection fluorescence microscopy, respectively. HFg increased permeability of the endothelial cell layer mainly through the transcellular pathway. While MβCD blocked Fg-increased transcellular and paracellular transport, myriocin affected only transcellular transport. Less pial venular leakage of albumin was observed in myriocin-treated HFg mice. HFg induced greater formation of functional caveolae, as indicated by colocalization of Na+-K+-ATPase with plasmalemmal vesicle-associated protein-1 by Förster resonance energy transfer and total internal reflection fluorescence microscopy. Our results suggest that elevated blood levels of Fg alter cerebrovascular permeability mainly by affecting caveolae-mediated transcytosis through modulation of de novo sphingolipid synthesis.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Syed Jalal Khundmiri
- Kidney Disease Program, Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Reeta Tyagi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Allison Gartung
- Department of Pathology, School of Medicine, Wayne State University, Detroit, Michigan
| | - William L Dean
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, Kentucky; and
| | - Menq-Jer Lee
- Department of Pathology, School of Medicine, Wayne State University, Detroit, Michigan
| | - David Lominadze
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky;
| |
Collapse
|
225
|
Walton BL, Getz TM, Bergmeier W, Lin FC, Uitte de Willige S, Wolberg AS. The fibrinogen γA/γ' isoform does not promote acute arterial thrombosis in mice. J Thromb Haemost 2014; 12:680-9. [PMID: 24916154 PMCID: PMC4098759 DOI: 10.1111/jth.12534] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 02/04/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND Elevated plasma fibrinogen is associated with arterial thrombosis in humans and promotes thrombosis in mice by increasing fibrin formation and thrombus fibrin content. Fibrinogen is composed of six polypeptide chains: (Aα, Bβ, and γ)2. Alternative splicing of the γ chain leads to a dominant form (γA/γA) and a minor species (γA/γ'). Epidemiological studies have detected elevated γA/γ' fibrinogen in patients with arterial thrombosis, suggesting that this isoform promotes thrombosis. However, in vitro data show that γA/γ' is anticoagulant due to its ability to sequester thrombin and suggest its expression is upregulated in response to inflammatory processes. OBJECTIVE To determine whether γA/γ' fibrinogen is prothrombotic in vivo. METHODS We separated γA/γA and γA/γ' fibrinogen from human plasma-purified fibrinogen and determined the effects on in vitro plasma clot formation and on in vivo thrombus formation and circulating thrombin-antithrombin complexes in mice. RESULTS AND CONCLUSIONS Both γA/γA and γA/γ' fibrinogen were cleaved by murine and human thrombin and were incorporated into murine and human clots. When γA/γA or γA/γ' was spiked into plasma, γA/γA increased the fibrin formation rate to a greater extent than γA/γ'. In mice, compared to controls, γA/γA infusion shortened the time to carotid artery occlusion, whereas γA/γ' infusion did not. Additionally, γA/γ' infusion led to lower levels of plasma thrombin-antithrombin complexes following arterial injury, whereas γA/γA infusion did not. These data suggest that γA/γ' binds thrombin in vivo and decreases prothrombotic activity. Together, these findings indicate that elevated levels of γA/γA fibrinogen promote arterial thrombosis in vivo, whereas γA/γ' does not.
Collapse
Affiliation(s)
- B L Walton
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | | | | | | |
Collapse
|
226
|
Hong LF, Li XL, Luo SH, Guo YL, Zhu CG, Qing P, Wu NQ, Li JJ. Association of fibrinogen with severity of stable coronary artery disease in patients with type 2 diabetic mellitus. DISEASE MARKERS 2014; 2014:485687. [PMID: 24803720 PMCID: PMC3997864 DOI: 10.1155/2014/485687] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 03/04/2014] [Accepted: 03/04/2014] [Indexed: 11/24/2022]
Abstract
BACKGROUND Some studies have suggested a relation of plasma fibrinogen to the severity of coronary artery disease (CAD). However, whether plasma fibrinogen can predict the presence and severity of CAD in patients with diabetes mellitus has not been determined. METHODS A total of consecutive 373 diabetic patients with typical angina pectoris who received coronary angiography were enrolled and classified into three groups by tertiles of Gensini score (GS, low group <8; intermediate group 8~28; high group >28). The relationship between fibrinogen and GS was evaluated. RESULTS There were correlations of fibrinogen with hemoglobin A1c, C-reactive protein, and GS (r = 0.17, r = 0.52, and r = 0.21, resp.; all P < 0.001). Area under the receivers operating characteristic curve of fibrinogen was 0.62 (95% CI 0.56-0.68, P < 0.001) for predicting a high GS. Multivariate analysis suggested that plasma fibrinogen was an independent predictor of a high GS for diabetic patients (OR = 1.40, 95% CI 1.04-1.88, and P = 0.026) after adjusting for traditional risk factors of CAD. CONCLUSIONS The present data indicated that plasma fibrinogen, a readily measurable systematic inflammatory marker, appeared to be an independent predictor for the severity of CAD in diabetic patients.
Collapse
Affiliation(s)
- Li-Feng Hong
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing 100037, China
- Department of Cardiology, The Fifth Hospital of Wuhan & Affiliated Guangci Hospital of Wuhan University, Wuhan 430050, China
| | - Xiao-Lin Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing 100037, China
| | - Song-Hui Luo
- Department of Cardiology, The Fifth Hospital of Wuhan & Affiliated Guangci Hospital of Wuhan University, Wuhan 430050, China
| | - Yuan-Lin Guo
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing 100037, China
| | - Cheng-Gang Zhu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing 100037, China
| | - Ping Qing
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing 100037, China
| | - Na-Qiong Wu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing 100037, China
| | - Jian-Jun Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing 100037, China
| |
Collapse
|
227
|
Söderholm M, Zia E, Hedblad B, Engström G. Leukocyte count and incidence of subarachnoid haemorrhage: a prospective cohort study. BMC Neurol 2014; 14:71. [PMID: 24708536 PMCID: PMC4234394 DOI: 10.1186/1471-2377-14-71] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 04/01/2014] [Indexed: 02/04/2023] Open
Abstract
Background Subarachnoid haemorrhage (SAH) is a devastating disease, in the majority of cases caused by a rupture of an arterial intracranial aneurysm. The effect of systemic low-grade inflammation on incidence of SAH is not known. The purpose of this study was to evaluate the relationship between leukocyte count, a marker of systemic inflammation, and incidence of SAH in a large cohort study. Methods Leukocyte count and other cardiovascular risk factors were measured in 19,794 individuals (17,083 men and 2,711 women, mean age 44 years) participating in a health screening program between 1974 and 1981. Incidence of SAH in relation to baseline leukocyte concentration was studied during a mean follow-up of 27 years in participants free from previous stroke. Results Ninety-five participants had a SAH, corresponding to an incidence of 22 per 100,000 in women and 17 per 100,000 in men. The hazard ratio for SAH per one standard deviation (2.01 × 109 cells/L) increase of leukocyte concentration was 1.26 (95% CI 1.05-1.53, p = 0.014) after adjustment for several potential confounding factors including smoking. In sensitivity analysis, there was a significant association in smokers but not in non-smokers. Conclusions High leukocyte count at baseline was associated with increased incidence of SAH, although this relationship might be restricted to smokers. The results support the view that low-grade systemic inflammation could be involved in the pathogenesis of SAH, or constitute an early risk marker for the disease.
Collapse
Affiliation(s)
- Martin Söderholm
- Cardiovascular Epidemiology Research Group, Department of Clinical Sciences Malmö, Lund University, CRC building 60 floor 13, Jan Waldenströms gata 35, 20502 Malmö, Sweden.
| | | | | | | |
Collapse
|
228
|
Ross Eckard A, Longenecker CT, Jiang Y, Debanne SM, Labbato D, Storer N, McComsey GA. Lipoprotein-associated phospholipase A2 and cardiovascular disease risk in HIV infection. HIV Med 2014; 15:537-46. [PMID: 24650269 DOI: 10.1111/hiv.12143] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2014] [Indexed: 12/25/2022]
Abstract
OBJECTIVES HIV-infected patients on antiretroviral therapy (ART) have an increased cardiovascular disease (CVD) risk as a result of heightened inflammation and immune activation, despite at times having normal lipids and few traditional risk factors. Biomarkers are needed to identify such patients before a clinical event. Lipoprotein-associated phospholipase A2 (Lp-PLA2 ) predicts CVD events in the general population. This study investigated the relationship between Lp-PLA2 and markers of CVD risk, systemic inflammation, immune activation, and coagulation in HIV infection. METHODS One hundred subjects on stable ART with normal fasting low-density lipoprotein (LDL) cholesterol were enrolled in the study. Plasma Lp-PLA2 concentrations were measured by enzyme-linked immunosorbent assay (ELISA; > 200 ng/mL was considered high CVD risk). Subclinical atherosclerosis, endothelial function, inflammation, immune activation and fasting lipids were also evaluated. RESULTS The median age of the patients was 47 years and 77% were male. Median (range) Lp-PLA2 was 209 (71-402) ng/mL. Fifty-seven per cent of patients had Lp-PLA2 concentrations > 200 ng/mL. Lp-PLA2 was positively correlated with soluble markers of inflammation or immune activation (tumour necrosis factor receptor-II, intercellular and vascular cellular adhesion molecules, and CD14; all R = 0.3; P < 0.01), and negatively correlated with coagulation markers (D-dimer and fibrinogen; both R = -0.2; P < 0.04). Lp-PLA2 was not correlated with lipids, coronary artery calcium score, or flow-mediated vasodilation, but trended towards a significant correlation with carotid intima-media thickness (R = 0.2; P = 0.05). CONCLUSIONS In this population with stable ART and normal LDL cholesterol, Lp-PLA2 was in the high CVD risk category in the majority of subjects. Lp-PLA2 appears to be associated with inflammation/immune activation, but also with anti-thrombotic effects. Lp-PLA2 may represent a valuable early biomarker of CVD risk in HIV infection before subclinical atherosclerosis can be detected.
Collapse
Affiliation(s)
- A Ross Eckard
- Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | | | | | | | | | | | | |
Collapse
|
229
|
von Frankenberg AD, do Nascimento FV, Gatelli LE, Nedel BL, Garcia SP, de Oliveira CSV, Saddi-Rosa P, Reis AF, Canani LH, Gerchman F. Major components of metabolic syndrome and adiponectin levels: a cross-sectional study. Diabetol Metab Syndr 2014; 6:26. [PMID: 24568287 PMCID: PMC3941563 DOI: 10.1186/1758-5996-6-26] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 02/18/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Adiponectin is a major regulator of glucose and lipid homeostasis by its insulin sensitizer properties. Since decreased insulin sensitivity is linked to metabolic syndrome (MS), decreased adiponectin levels may be related to its development. The purpose of the study was to investigate the relationship between adiponectin levels and MS. METHODS Firstly, we cross-sectionally examined subjects with or without MS submitted to an oral glucose tolerance test at Hospital de Clínicas de Porto Alegre (n = 172). A replication analysis was performed in subjects (n = 422) undergoing cardiac angiography at Hospital São Paulo. Subchronic inflammation (US-CRP), coagulation marker (fibrinogen), insulin sensitivity and resistance (Matsuda ISI and HOMA-IR) were estimated. Plasma total and high molecular weight (HMW) adiponectin were measured. RESULTS Total and HMW adiponectin levels were lower in MS subjects (P < 0.05). Total adiponectin levels were lower in the presence of high waist circumference, low HDL-cholesterol and elevated triglyceride criteria in both samples and by elevated blood pressure and glucose criteria in Porto Alegre. HMW adiponectin levels were lower in the presence of low HDL-cholesterol, elevated triglycerides, and glucose criteria. Total adiponectin levels were positively related with HDL-cholesterol and ISI Matsuda, negatively related with waist circumference, glucose, triglycerides, HOMA-IR, and US-CRP and not related with blood pressure. While adjusting for sex and age, increased adiponectin levels remained associated with a reduced prevalence ratio for MS in both cohorts (P = 0.001). CONCLUSIONS Adiponectin levels decreased with increasing number of MS criteria, and it is in part determined by its relationship with HDL, triglycerides and abdominal adiposity.
Collapse
Affiliation(s)
- Anize D von Frankenberg
- Post-Graduate Endocrinology Program, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Hospital de Clínicas de Porto Alegre. Rua Ramiro Barcelos, 2350 – Prédio 12. 4° andar, 90035-003 Porto Alegre, RS, Brazil
| | - Filipe V do Nascimento
- Post-Graduate Endocrinology Program, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Lucas Eduardo Gatelli
- Post-Graduate Endocrinology Program, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Bárbara L Nedel
- Post-Graduate Endocrinology Program, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Sheila P Garcia
- Post-Graduate Endocrinology Program, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | | | - Pedro Saddi-Rosa
- Endocrinology Unit, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - André F Reis
- Endocrinology Unit, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Luis H Canani
- Post-Graduate Endocrinology Program, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Metabolism Unit, Endocrinology Division, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Fernando Gerchman
- Post-Graduate Endocrinology Program, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Metabolism Unit, Endocrinology Division, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| |
Collapse
|
230
|
Su JJ, Park SK, Hsieh TM. The Effect of Testosterone on Cardiovascular Disease. Am J Mens Health 2014; 8:470-91. [DOI: 10.1177/1557988314522642] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular disease is the leading cause of death in the United States. Testosterone is the principal male sex hormone and plays an important role in men’s health and well-being. Historically, testosterone was believed to adversely affect cardiovascular function. However, contemporary literature has refuted this traditional thinking; testosterone has been suggested to have a protective effect on cardiovascular function through its effects on the vascular system. Data from modern research indicate that hypogonadism is closely related to the development of various cardiovascular risk factors, including hyperlipidemia and insulin resistance. Several studies have demonstrated beneficial effects of testosterone supplementation therapy on reversing symptoms of hypogonadism and improving cardiovascular disease risk profiles. In this review, we perform a critical analysis on the association between testosterone and cardiovascular disease.
Collapse
|
231
|
Severity of obstructive sleep apnea is associated with elevated plasma fibrinogen in otherwise healthy patients. Sleep Breath 2014; 18:761-6. [PMID: 24510497 DOI: 10.1007/s11325-014-0938-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 12/09/2013] [Accepted: 01/09/2014] [Indexed: 02/07/2023]
Abstract
PURPOSE Obstructive sleep apnea (OSA) has been implicated in both cardiovascular and cerebrovascular diseases. Systemic inflammation and coagulation may be related to cardiovascular pathophysiology in patients with OSA. Fibrinogen is a major coagulation protein associated with inflammation, and long-term elevated plasma fibrinogen is associated with an increased risk of major cardiovascular diseases. We assessed whether severity of OSA is associated with levels of fibrinogen in newly diagnosed, untreated, and otherwise healthy OSA patients. METHODS We studied 36 men with OSA and 18 male control subjects (apnea-hypopnea index [AHI]<5 events/h). OSA patients were divided into mild (AHI≥5<15 events/h) and severe (AHI≥15 events/h) OSA groups. Morning fibrinogen levels in OSA patients were compared to those in control subjects of similar age, body mass index, blood pressure, smoking habits, and alcohol consumption. RESULTS Fibrinogen levels were significantly elevated in patients with severe OSA compared to both control (P=0.003) and mild OSA (P=0.02) subjects after adjustment for covariates. However, there were no significant differences in fibrinogen levels between mild OSA and control subjects. Fibrinogen levels were directly related to AHI and arousal index and inversely related to mean and lowest oxygen saturation during sleep. CONCLUSIONS Severity of OSA was associated with increased fibrinogen level independent of other factors, suggesting that apneic events and oxygen desaturation during sleep are mechanisms for increased fibrinogen levels in patients with OSA.
Collapse
|
232
|
Eckard AR, Jiang Y, Debanne SM, Funderburg NT, McComsey GA. Effect of 24 weeks of statin therapy on systemic and vascular inflammation in HIV-infected subjects receiving antiretroviral therapy. J Infect Dis 2014; 209:1156-64. [PMID: 24415784 DOI: 10.1093/infdis/jiu012] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Human immunodeficiency virus (HIV)-infected individuals are at increased risk of cardiovascular disease (CVD) due in part to inflammation. Statins decrease inflammation in the general population, but their effect during HIV infection is largely unknown. METHODS This is an ongoing randomized, double-blinded, placebo-controlled trial to evaluate the effect of statin therapy on inflammatory markers during HIV infection. Subjects received rosuvastatin 10 mg daily or placebo for 24 weeks. Subjects were receiving stable (>12 weeks) antiretroviral therapy and had a low-density lipoprotein (LDL) cholesterol level of ≤130 mg/dL and evidence of heightened immune activation or inflammation. This was a prespecified interim analysis. RESULTS A total of 147 subjects were enrolled (78% were male, 70% were black, and the median age was 47 years). By 24 weeks, LDL cholesterol levels had decreased in the statin group, compared with an increase in the placebo group (-28% vs +3.8%; P < .01). A 10% reduction in the lipoprotein-associated phospholipase A2 (Lp-PLA2) level was seen in the statin group, compared with a 2% reduction in the placebo group (P < .01). In multivariable regression, receipt of statin treatment and having a nadir CD4(+) T-cell count of ≤100 cell/µL were the only statistically significant predictors of a decrease in Lp-PLA2 level. Markers of systemic inflammation did not change significantly between groups. CONCLUSIONS Twenty-four weeks of rosuvastatin therapy significantly decreased the level of Lp-PLA2, a vascular-specific, inflammatory enzyme that predicts cardiovascular events in the general population. Statins may hold promise as a means of attenuating CVD risk in HIV-infected individuals by decreasing Lp-PLA2 levels.
Collapse
|
233
|
Genetic polymorphism of β-fibrinogen gene-455G/A can contribute to the risk of ischemic stroke. Neurol Sci 2013; 35:151-61. [PMID: 24366241 DOI: 10.1007/s10072-013-1608-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 12/11/2013] [Indexed: 10/25/2022]
Abstract
Many studies have investigated the association between the β-fibrinogen gene-455G/A (FGβ-455G/A) polymorphism and the risk of ischemic stroke. However, these evidences were inadequate to provide stronger conclusions because most studies were generally small. To shed light on these inconclusive findings, we conducted a large sample size meta-analysis of studies relating to the FGβ-455G/A polymorphism and the risk of ischemic stroke. Odds ratios with a 95 % confidence interval were used to investigate the association between FGβ-455G/A polymorphism and ischemic stroke. Publication bias was tested by Egger's test and funnel plot. Inconsistency index and Cochran's Q statistic were used to check heterogeneity. Cumulative and recursive cumulative meta-analyses were performed to provide a framework for updating a genetic effect from all of the included studies. Twenty-six independent publications with 4,070 cases and 4,649 controls were included in this meta-analysis. Results showed that the β-fibrinogen-455G/A polymorphism was significantly associated with the risk of ischemic stroke. The FGβ-455G/A polymorphism was found to be a risk factor for ischemic stroke in Asians and adults, while association was not observed for Caucasians and juveniles based on the small size and it may be necessary to conduct larger studies on them to investigate the association in the future. The cumulative meta-analysis indicated a decline from 1998 to 2003, and the results remained stable during the period 2004-2012. The results indicate that FGβ-455G/A polymorphism may be a susceptible predictor of ischemic stroke. More studies are needed to elucidate the relationship further.
Collapse
|
234
|
Hsu DC, Sereti I, Ananworanich J. Serious Non-AIDS events: Immunopathogenesis and interventional strategies. AIDS Res Ther 2013; 10:29. [PMID: 24330529 PMCID: PMC3874658 DOI: 10.1186/1742-6405-10-29] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 11/26/2013] [Indexed: 12/14/2022] Open
Abstract
Despite the major advances in the management of HIV infection, HIV-infected patients still have greater morbidity and mortality than the general population. Serious non-AIDS events (SNAEs), including non-AIDS malignancies, cardiovascular events, renal and hepatic disease, bone disorders and neurocognitive impairment, have become the major causes of morbidity and mortality in the antiretroviral therapy (ART) era. SNAEs occur at the rate of 1 to 2 per 100 person-years of follow-up. The pathogenesis of SNAEs is multifactorial and includes the direct effect of HIV and associated immunodeficiency, underlying co-infections and co-morbidities, immune activation with associated inflammation and coagulopathy as well as ART toxicities. A number of novel strategies such as ART intensification, treatment of co-infection, the use of anti-inflammatory drugs and agents that reduce microbial translocation are currently being examined for their potential effects in reducing immune activation and SNAEs. However, currently, initiation of ART before advanced immunodeficiency, smoking cessation, optimisation of cardiovascular risk factors and treatment of HCV infection are most strongly linked with reduced risk of SNAEs or mortality. Clinicians should therefore focus their attention on addressing these issues prior to the availability of further data.
Collapse
|
235
|
Masoudkabir F, Sarrafzadegan N, Eisenberg MJ. Effects of opium consumption on cardiometabolic diseases. Nat Rev Cardiol 2013; 10:733-40. [PMID: 24145895 DOI: 10.1038/nrcardio.2013.159] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Opium is the second-most-commonly abused substance (after tobacco) in developing countries of the Middle East region, and in many Asian nations. One of the reasons for the high prevalence of opium abuse in these countries is a traditional belief among Eastern people, even including some medical staff, that opium might have beneficial effects on cardiovascular health and in the control of diabetes mellitus, hypertension, and dyslipidaemia. In this Perspectives article, we summarize the current understanding of the pharmacotoxicology of opium and its specific effects on glycaemic control, blood pressure, lipid profile, and atherosclerosis. On the basis of the available evidence, we believe not only that opium has no ameliorating effect on cardiovascular diseases, but also that the use of this drug might have adverse effects on these conditions. Therefore, people should be educated about the hazardous effects of opium consumption on cardiometabolic diseases.
Collapse
Affiliation(s)
- Farzad Masoudkabir
- Department of Cardiology, Tehran Heart Center, Tehran University of Medical Sciences, Karegar Shomali Street, Jalal al-Ahmad Cross, 14117-13138, Tehran, Iran
| | | | | |
Collapse
|
236
|
Background Diet and Fat Type Alters Plasma Lipoprotein Response but not Aortic Cholesterol Accumulation in F1B Golden Syrian Hamsters. Lipids 2013; 48:1177-84. [DOI: 10.1007/s11745-013-3840-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 08/15/2013] [Indexed: 11/26/2022]
|
237
|
Abstract
Traumatic brain injury (TBI) has been associated with various neurological disorders. However, the role of cerebrovascular dysfunction and its mechanisms associated with TBI are still not well understood. Inflammation is the main cause of vascular dysfunction. It affects properties of blood components and the vascular wall leading to changes in blood flow and in interaction of blood components and vascular endothelium exacerbating microcirculatory complications during inflammatory diseases. One of the markers of inflammation is a plasma adhesion protein, fibrinogen (Fg). At elevated levels, Fg can also cause inflammatory responses. One of the manifestations of inflammatory responses is an increase in microvascular permeability leading to accumulation of plasma proteins in the subendothelial matrix and causing vascular remodelling. This has a most devastating effect on cerebral circulation after TBI that is accompanied with an elevation of plasma level of Fg and with an increased cerebrovascular permeability in injury penumbra impairing the normal healing process. This study reviews cerebrovascular alterations after TBI, considers the consequences of increased blood-brain barrier permeability, defines the role of elevated level of Fg and discusses the potential mechanisms of its action leading to vascular dysfunction, which subsequently can cause impairment in neuronal function. Thus, possible mechanisms of vasculo-neuronal dysfunction after TBI are considered.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology and Biophysics, University of Louisville, School of Medicine , Louisville, KY , USA
| | | |
Collapse
|
238
|
Zhang X, Johnson AD, Hendricks AE, Hwang SJ, Tanriverdi K, Ganesh SK, Smith NL, Peyser PA, Freedman JE, O'Donnell CJ. Genetic associations with expression for genes implicated in GWAS studies for atherosclerotic cardiovascular disease and blood phenotypes. Hum Mol Genet 2013; 23:782-95. [PMID: 24057673 DOI: 10.1093/hmg/ddt461] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Genome-wide association studies (GWAS) have uncovered many genetic associations for cardiovascular disease (CVD). However, data are limited regarding causal genetic variants within implicated loci. We sought to identify regulatory variants (cis- and trans-eQTLs) affecting expression levels of 93 genes selected by their proximity to SNPs with significant associations in prior GWAS for CVD traits. Expression levels were measured by qRT-PCR in leukocytes from 1846 Framingham Heart Study participants. An additive genetic model was applied to 2.5 million imputed SNPs for each gene. Approximately 45% of genes (N = 38) harbored at least one cis-eSNP after a regional multiple-test adjustment. Applying a more rigorous significance threshold (P < 5 × 10(-8)), we found the expression level of 10 genes was significantly associated with more than one cis-eSNP. The top cis-eSNPs for 7 of these 10 genes exhibited moderate-to-strong association with ≥ 1 CVD clinical phenotypes. Several eSNPs or proxy SNPs (r(2) = 1) were replicated by other eQTL studies. After adjusting for the lead GWAS SNPs for the 10 genes, expression variances explained by top cis-eSNPs were attenuated markedly for LPL, FADS2 and C6orf184, suggesting a shared genetic basis for the GWAS and expression trait. A significant association between cis-eSNPs, gene expression and lipid levels was discovered for LPL and C6orf184. In conclusion, strong cis-acting variants are localized within nearly half of the GWAS loci studied, with particularly strong evidence for a regulatory role of the top GWAS SNP for expression of LPL, FADS2 and C6orf184.
Collapse
Affiliation(s)
- Xiaoling Zhang
- Division of Intramural Research, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
239
|
Sabater-Lleal M, Huang J, Chasman D, Naitza S, Dehghan A, Johnson AD, Teumer A, Reiner AP, Folkersen L, Basu S, Rudnicka AR, Trompet S, Mälarstig A, Baumert J, Bis JC, Guo X, Hottenga JJ, Shin SY, Lopez LM, Lahti J, Tanaka T, Yanek LR, Oudot-Mellakh T, Wilson JF, Navarro P, Huffman JE, Zemunik T, Redline S, Mehra R, Pulanic D, Rudan I, Wright AF, Kolcic I, Polasek O, Wild SH, Campbell H, Curb JD, Wallace R, Liu S, Eaton CB, Becker DM, Becker LC, Bandinelli S, Räikkönen K, Widen E, Palotie A, Fornage M, Green D, Gross M, Davies G, Harris SE, Liewald DC, Starr JM, Williams FM, Grant P, Spector TD, Strawbridge RJ, Silveira A, Sennblad B, Rivadeneira F, Uitterlinden AG, Franco OH, Hofman A, van Dongen J, Willemsen G, Boomsma DI, Yao J, Jenny NS, Haritunians T, McKnight B, Lumley T, Taylor KD, Rotter JI, Psaty BM, Peters A, Gieger C, Illig T, Grotevendt A, Homuth G, Völzke H, Kocher T, Goel A, Franzosi MG, Seedorf U, Clarke R, Steri M, Tarasov KV, Sanna S, Schlessinger D, Stott DJ, Sattar N, Buckley BM, Rumley A, Lowe GD, McArdle WL, Chen MH, Tofler GH, Song J, Boerwinkle E, Folsom AR, et alSabater-Lleal M, Huang J, Chasman D, Naitza S, Dehghan A, Johnson AD, Teumer A, Reiner AP, Folkersen L, Basu S, Rudnicka AR, Trompet S, Mälarstig A, Baumert J, Bis JC, Guo X, Hottenga JJ, Shin SY, Lopez LM, Lahti J, Tanaka T, Yanek LR, Oudot-Mellakh T, Wilson JF, Navarro P, Huffman JE, Zemunik T, Redline S, Mehra R, Pulanic D, Rudan I, Wright AF, Kolcic I, Polasek O, Wild SH, Campbell H, Curb JD, Wallace R, Liu S, Eaton CB, Becker DM, Becker LC, Bandinelli S, Räikkönen K, Widen E, Palotie A, Fornage M, Green D, Gross M, Davies G, Harris SE, Liewald DC, Starr JM, Williams FM, Grant P, Spector TD, Strawbridge RJ, Silveira A, Sennblad B, Rivadeneira F, Uitterlinden AG, Franco OH, Hofman A, van Dongen J, Willemsen G, Boomsma DI, Yao J, Jenny NS, Haritunians T, McKnight B, Lumley T, Taylor KD, Rotter JI, Psaty BM, Peters A, Gieger C, Illig T, Grotevendt A, Homuth G, Völzke H, Kocher T, Goel A, Franzosi MG, Seedorf U, Clarke R, Steri M, Tarasov KV, Sanna S, Schlessinger D, Stott DJ, Sattar N, Buckley BM, Rumley A, Lowe GD, McArdle WL, Chen MH, Tofler GH, Song J, Boerwinkle E, Folsom AR, Rose LM, Franco-Cereceda A, Teichert M, Ikram MA, Mosley TH, Bevan S, Dichgans M, Rothwell PM, Sudlow CLM, Hopewell JC, Chambers JC, Saleheen D, Kooner JS, Danesh J, Nelson CP, Erdmann J, Reilly MP, Kathiresan S, Schunkert H, Morange PE, Ferrucci L, Eriksson JG, Jacobs D, Deary IJ, Soranzo N, Witteman JCM, de Geus EJC, Tracy RP, Hayward C, Koenig W, Cucca F, Jukema JW, Eriksson P, Seshadri S, Markus HS, Watkins H, Samani NJ, VTE consortium, STROKE Consortium, Wellcome Trust Case Control Consortium 2 (WTCCC2), C4D consortium, CARDIoGRAM consortium, Wallaschofski H, Smith NL, Tregouet D, Ridker PM, Tang W, Strachan DP, Hamsten A, O’Donnell CJ. Multiethnic meta-analysis of genome-wide association studies in >100 000 subjects identifies 23 fibrinogen-associated Loci but no strong evidence of a causal association between circulating fibrinogen and cardiovascular disease. Circulation 2013; 128:1310-24. [PMID: 23969696 PMCID: PMC3842025 DOI: 10.1161/circulationaha.113.002251] [Show More Authors] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 07/12/2013] [Indexed: 11/16/2022]
Abstract
BACKGROUND Estimates of the heritability of plasma fibrinogen concentration, an established predictor of cardiovascular disease, range from 34% to 50%. Genetic variants so far identified by genome-wide association studies explain only a small proportion (<2%) of its variation. METHODS AND RESULTS We conducted a meta-analysis of 28 genome-wide association studies including >90 000 subjects of European ancestry, the first genome-wide association meta-analysis of fibrinogen levels in 7 studies in blacks totaling 8289 samples, and a genome-wide association study in Hispanics totaling 1366 samples. Evaluation for association of single-nucleotide polymorphisms with clinical outcomes included a total of 40 695 cases and 85 582 controls for coronary artery disease, 4752 cases and 24 030 controls for stroke, and 3208 cases and 46 167 controls for venous thromboembolism. Overall, we identified 24 genome-wide significant (P<5×10(-8)) independent signals in 23 loci, including 15 novel associations, together accounting for 3.7% of plasma fibrinogen variation. Gene-set enrichment analysis highlighted key roles in fibrinogen regulation for the 3 structural fibrinogen genes and pathways related to inflammation, adipocytokines, and thyrotrophin-releasing hormone signaling. Whereas lead single-nucleotide polymorphisms in a few loci were significantly associated with coronary artery disease, the combined effect of all 24 fibrinogen-associated lead single-nucleotide polymorphisms was not significant for coronary artery disease, stroke, or venous thromboembolism. CONCLUSIONS We identify 23 robustly associated fibrinogen loci, 15 of which are new. Clinical outcome analysis of these loci does not support a causal relationship between circulating levels of fibrinogen and coronary artery disease, stroke, or venous thromboembolism.
Collapse
|
240
|
Kaptoge S, Seshasai SRK, Gao P, Freitag DF, Butterworth AS, Borglykke A, Di Angelantonio E, Gudnason V, Rumley A, Lowe GDO, Jørgensen T, Danesh J. Inflammatory cytokines and risk of coronary heart disease: new prospective study and updated meta-analysis. Eur Heart J 2013; 35:578-89. [PMID: 24026779 DOI: 10.1093/eurheartj/eht367] [Citation(s) in RCA: 489] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIMS Because low-grade inflammation may play a role in the pathogenesis of coronary heart disease (CHD), and pro-inflammatory cytokines govern inflammatory cascades, this study aimed to assess the associations of several pro-inflammatory cytokines and CHD risk in a new prospective study, including meta-analysis of prospective studies. METHODS AND RESULTS Interleukin-6 (IL-6), IL-18, matrix metalloproteinase-9 (MMP-9), soluble CD40 ligand (sCD40L), and tumour necrosis factor-α (TNF-α) were measured at baseline in a case-cohort study of 1514 participants and 833 incident CHD events within population-based prospective cohorts at the Danish Research Centre for Prevention and Health. Age- and sex-adjusted hazard ratios (HRs) for CHD per 1-SD higher log-transformed baseline levels were: 1.37 (95% CI: 1.21-1.54) for IL-6, 1.26 (1.11-1.44) for IL-18, 1.30 (1.16-1.46) for MMP-9, 1.01 (0.89-1.15) for sCD40L, and 1.13 (1.01-1.27) for TNF-α. Multivariable adjustment for conventional vascular risk factors attenuated the HRs to: 1.26 (1.08-1.46) for IL-6, 1.12 (0.95-1.31) for IL-18, 1.21 (1.05-1.39) for MMP-9, 0.93 (0.78-1.11) for sCD40L, and 1.14 (1.00-1.31) for TNF-α. In meta-analysis of up to 29 population-based prospective studies, adjusted relative risks for non-fatal MI or CHD death per 1-SD higher levels were: 1.25 (1.19-1.32) for IL-6; 1.13 (1.05-1.20) for IL-18; 1.07 (0.97-1.19) for MMP-9; 1.07 (0.95-1.21) for sCD40L; and 1.17 (1.09-1.25) for TNF-α. CONCLUSIONS Several different pro-inflammatory cytokines are each associated with CHD risk independent of conventional risk factors and in an approximately log-linear manner. The findings lend support to the inflammation hypothesis in vascular disease, but further studies are needed to assess causality.
Collapse
Affiliation(s)
- Stephen Kaptoge
- Strangeways Research Laboratory, Department of Public Health and Primary Care, University of Cambridge, Worts Causeway, CB1 8RN Cambridge, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
241
|
Shibata Y, Abe S, Inoue S, Igarashi A, Yamauchi K, Aida Y, Kishi H, Nunomiya K, Nakano H, Sato M, Sato K, Kimura T, Nemoto T, Watanabe T, Konta T, Ueno Y, Kato T, Kayama T, Kubota I. Relationship between plasma fibrinogen levels and pulmonary function in the japanese population: the Takahata study. Int J Med Sci 2013; 10:1530-6. [PMID: 24046528 PMCID: PMC3775111 DOI: 10.7150/ijms.7256] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 08/16/2013] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Plasma fibrinogen is considered a biomarker of respiratory disease, owing to the relationship between plasma fibrinogen and pulmonary function established in Western populations. However, such a relationship has not yet been confirmed in an Asian population. We assessed this relationship in the general Japanese population. METHODS Totally, 3,257 men and women aged ≥40 years who participated in a community-based annual health checkup in Takahata, Japan, from 2004 to 2006, underwent spirometry, and their plasma fibrinogen levels were determined. RESULTS We found an inverse relationship between spirometric measures (percent predicted forced vital capacity [%FVC] and forced expiratory volume in 1s [%FEV1], and FEV1/FVC) and plasma fibrinogen levels in men, but not in women. The plasma fibrinogen levels were significantly higher in subjects with restrictive, obstructive, and mixed ventilatory disorders than in those with normal spirometry results. Multiple linear regression analysis revealed that in men, plasma fibrinogen levels were predictive for %FVC and %FEV1 (independent of age, body mass index, and cigarette smoking) but not for FEV1/FVC. CONCLUSIONS Plasma fibrinogen was significantly associated with pulmonary function in Japanese men, and as such, plasma fibrinogen might be a potent biomarker for pulmonary dysfunction in men.
Collapse
Affiliation(s)
- Yoko Shibata
- 1. Department of Cardiology, Pulmonology, and Nephrology
| | - Shuichi Abe
- 1. Department of Cardiology, Pulmonology, and Nephrology
| | - Sumito Inoue
- 1. Department of Cardiology, Pulmonology, and Nephrology
| | - Akira Igarashi
- 1. Department of Cardiology, Pulmonology, and Nephrology
| | - Keiko Yamauchi
- 1. Department of Cardiology, Pulmonology, and Nephrology
| | - Yasuko Aida
- 1. Department of Cardiology, Pulmonology, and Nephrology
| | - Hiroyuki Kishi
- 1. Department of Cardiology, Pulmonology, and Nephrology
| | - Keiko Nunomiya
- 1. Department of Cardiology, Pulmonology, and Nephrology
| | - Hiroshi Nakano
- 1. Department of Cardiology, Pulmonology, and Nephrology
| | - Masamichi Sato
- 1. Department of Cardiology, Pulmonology, and Nephrology
| | - Kento Sato
- 1. Department of Cardiology, Pulmonology, and Nephrology
| | - Tomomi Kimura
- 1. Department of Cardiology, Pulmonology, and Nephrology
| | - Takako Nemoto
- 1. Department of Cardiology, Pulmonology, and Nephrology
| | - Tetsu Watanabe
- 1. Department of Cardiology, Pulmonology, and Nephrology
| | - Tsuneo Konta
- 1. Department of Cardiology, Pulmonology, and Nephrology
| | - Yoshiyuki Ueno
- 2. Global Center of Excellence Program Study Group, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Takeo Kato
- 2. Global Center of Excellence Program Study Group, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Takamasa Kayama
- 2. Global Center of Excellence Program Study Group, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Isao Kubota
- 1. Department of Cardiology, Pulmonology, and Nephrology
| |
Collapse
|
242
|
Mayhew PD, Savigny MR, Otto CM, Brown DC, Brooks MB, Bentley AM, Runge JJ, Callan MB. Evaluation of coagulation in dogs with partial or complete extrahepatic biliary tract obstruction by means of thromboelastography. J Am Vet Med Assoc 2013; 242:778-85. [PMID: 23445288 DOI: 10.2460/javma.242.6.778] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To characterize in vitro coagulation status in a cohort of dogs with extrahepatic biliary tract obstruction (EHBO) and to evaluate these patients for hypercoagulability by means of thromboelastography. DESIGN Prospective cohort study. Animals-10 dogs with EHBO and 19 healthy control dogs. PROCEDURES Partial or complete EHBO was confirmed via exploratory celiotomy. Venous blood samples were collected for evaluation of prothrombin time (PT) and activated partial thromboplastin time (APTT); fibrinogen and D-dimer concentrations; protein C and antithrombin activities; and factor VII, VIII, and XI coagulant activities in plasma as well as thromboelastography in whole blood. Thromboelastography variables were measured from the thromboelastography tracing, and a coagulation index was calculated. Thromboelastography results were compared with those of healthy control dogs previously evaluated by the same laboratory. RESULTS Hypercoagulability was diagnosed in all dogs with EHBO on the basis of a high coagulation index. Thromboelastography variables, including maximal amplitude, α-angle, and coagulation index, were significantly higher, and K (clot formation time) and R (reaction time) were significantly lower in these dogs than in control dogs. All dogs with EHBO had PT and APTT within respective reference ranges. Plasma D-dimer and fibrinogen concentrations were above reference ranges in 8 and 7 dogs, respectively, and protein C and antithrombin activities were below reference ranges in 3 and 1 dogs, respectively. CONCLUSIONS AND CLINICAL RELEVANCE In vitro hypercoagulability was commonly detected in dogs with naturally occurring EHBO. The traditional view of EHBO as a disease that causes hypocoagulability may need to be reconsidered.
Collapse
Affiliation(s)
- Philipp D Mayhew
- Department of Clinical Studies-Philadelphia, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | |
Collapse
|
243
|
DNA methylation profiling of the fibrinogen gene landscape in human cells and during mouse and zebrafish development. PLoS One 2013; 8:e73089. [PMID: 23991173 PMCID: PMC3749180 DOI: 10.1371/journal.pone.0073089] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 07/17/2013] [Indexed: 11/19/2022] Open
Abstract
The fibrinogen genes FGA, FGB and FGG show coordinated expression in hepatocytes. Understanding the underlying transcriptional regulation may elucidate how their tissue-specific expression is maintained and explain the high variability in fibrinogen blood levels. DNA methylation of CpG-poor gene promoters is dynamic with low methylation correlating with tissue-specific gene expression but its direct effect on gene regulation as well as implications of non-promoter CpG methylation are not clear. Here we compared methylation of CpG sites throughout the fibrinogen gene cluster in human cells and mouse and zebrafish tissues. We observed low DNA methylation of the CpG-poor fibrinogen promoters and of additional regulatory elements (the liver enhancers CNC12 and PFE2) in fibrinogen-expressing samples. In a gene reporter assay, CpG-methylation in the FGA promoter reduced promoter activity, suggesting a repressive function for DNA methylation in the fibrinogen locus. In mouse and zebrafish livers we measured reductions in DNA methylation around fibrinogen genes during development that were preceded by increased fibrinogen expression and tri-methylation of Histone3 lysine4 (H3K4me3) in fibrinogen promoters. Our data support a model where changes in hepatic transcription factor expression and histone modification provide the switch for increased fibrinogen gene expression in the developing liver which is followed by reduction of CpG methylation.
Collapse
|
244
|
Harhay MO, Tracy RP, Bagiella E, Barr RG, Pinder D, Hundley WG, Bluemke DA, Kronmal RA, Lima JAC, Kawut SM. Relationship of CRP, IL-6, and fibrinogen with right ventricular structure and function: the MESA-Right Ventricle Study. Int J Cardiol 2013; 168:3818-24. [PMID: 23932860 DOI: 10.1016/j.ijcard.2013.06.028] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 06/03/2013] [Accepted: 06/20/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND/OBJECTIVES Inflammation contributes to the pathogenesis of disease associated with the left ventricle (LV); yet, our understanding of the effect of inflammation on the right ventricle (RV) is quite limited. METHODS AND RESULTS The relationships of C-reactive protein (CRP), interleukin-6 (IL-6) and fibrinogen with RV morphology and function (from cardiac MRI) were examined in participants free of clinical cardiovascular disease (n=4009) from the Multi-Ethnic Study of Atherosclerosis (MESA)-RV study. Multivariable regressions (linear, quantile [25th and 75th] and generalized additive models [GAM]) were used to examine the independent association of CRP, IL-6 and fibrinogen with RV mass, RV end-diastolic volume (RVEDV), RV end-systolic volume (RVESV), RV stroke volume (RVSV) and RV ejection fraction (RVEF). Unadjusted and adjusted analyses revealed strong inverse associations between both CRP and IL-6 with RV mass, RVEDV, RVESV and RVSV (all p<0.01); there were no associations with RVEF. These relationships remained significant after adjustment for the respective LV parameters and lung function. However, GAM models suggested that extreme values of CRP and IL-6 might have positive associations with RV parameters. Fibrinogen showed significant associations in unadjusted models, but no associations after adjustment or in sensitivity analyses. CONCLUSION Levels of CRP and IL-6 are independently associated with RV morphology even after adjustment for the respective LV measure in this multi-ethnic population free of clinical cardiovascular disease. Systemic inflammation may contribute to RV structural changes independent of effects on the LV.
Collapse
Affiliation(s)
- Michael O Harhay
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Teramoto T, Sasaki J, Ishibashi S, Birou S, Daida H, Dohi S, Egusa G, Hiro T, Hirobe K, Iida M, Kihara S, Kinoshita M, Maruyama C, Ohta T, Okamura T, Yamashita S, Yokode M, Yokote K. Cardiovascular disease risk factors other than dyslipidemia. Executive summary of the Japan Atherosclerosis Society (JAS) guidelines for the diagnosis and prevention of atherosclerotic cardiovascular diseases in Japan - 2012 version. J Atheroscler Thromb 2013; 20:733-42. [PMID: 23892529 DOI: 10.5551/jat.17368] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Tamio Teramoto
- Committee for Epidemiology and Clinical Management of Atherosclerosis
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
246
|
Carbayo Herencia JA, Simarro Rueda M, Artigao Ródenas LM, Divisón Garrote JA, Caldevilla Bernardo D, Ponce García I, Sanchis Domènech C. [Relationship between inflammation marker and all-cause and cardiovascular mortality in a prospective cohort study]. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2013; 25:56-62. [PMID: 23849212 DOI: 10.1016/j.arteri.2013.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 01/10/2013] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Inflammation is present in every stage of the atherosclerosis process, therefore, inflammation hallmarks such as the fibrinogen can be related to the complications in which it intervenes, mortality is one of them. The objective of this study is to assess the association of the fibrinogen with all-cause mortality in men from general population sample obtained by random sampling in the Spanish region of Albacete. METHODS A total of 506men without cardiovascular events with 10.6years (SD=2.3) of follow-up, volunteered to participate in a prospective cohort study. The assessment of the fibrinogen as a predictor variable has been calculated after adjusting it by age, hypertension, diabetes mellitus, obesity, total cholesterol, HDL-cholesterol/triglycerides ratio, and smoking habit applying a Cox regression model. The adjustment has been made by adding the fibrinogen to the model, as a qualitative variable (<400 and ≥400mg/dl). RESULTS The average age of the participants was 46.6years old (DE=16.8). After the adjustment, the hyperfibrinogenemia (≥400mg/dl) showed a hazard ratio (HR) for all-cause mortality of 1.85 (95%CI: 1.05-3.26) and for cardiovascular mortality HR=2.69 (95%CI: 1.09-6.63). CONCLUSIONS In men without cardiovascular events of our study, fibrinogen was showed as an independent predictor of all-cause mortality and cardiovascular mortality.
Collapse
|
247
|
Seventeen year risk of all-cause and cause-specific mortality associated with C-reactive protein, fibrinogen and leukocyte count in men and women: the EPIC-Norfolk study. Eur J Epidemiol 2013; 28:541-50. [PMID: 23821244 DOI: 10.1007/s10654-013-9819-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 06/20/2013] [Indexed: 12/25/2022]
Abstract
There is strong evidence from observational studies suggesting serum C-reactive protein (CRP) is associated with cardiovascular and all-cause mortality. However, less is known about whether there are differences in the association of CRP with all-cause or cause specific mortality by sex, smoking, body mass index (BMI), or physical activity. We aimed to investigate these interactions and also investigate and compare the association of CRP and other inflammation markers (i.e., fibrinogen and leukocyte count) with all-cause and cause-specific mortality. Men and women aged 40-79 were recruited in 1993-1997 in the EPIC-Norfolk cohort study. A total of 16,850 participants with high-sensitivity assayed CRP data who had no known cancer, myocardial infarction and stroke at baseline were entered in the analysis to test the association of CRP, fibrinogen and leukocyte count with risk of all-cause and cause specific mortality. A total of, 2,603 all-cause deaths (1,452 in men) including 823 cardiovascular and 1,035 cancer deaths, were observed after 231,000 person-years of follow-up (median 14.3 years). CRP was positively associated with risk of all-cause, cardiovascular, and non-cancer non-cardiovascular mortality independent of established risk factors. The hazard ratio of all-cause mortality (95 % CI) for participants with CRP in the range of 3-10 and >10 mg/l (vs. <0.5 mg/l) was 1.56 (1.26-1.93) and 1.87 (1.43-2.43) respectively in men and 1.34 (1.07-1.68) and 1.98 (1.50-2.63) in women. The association was less positively graded in women with the increased risk being significant only at higher levels of the CRP distribution. The association persisted in never smokers and did not vary by levels of BMI or physical activity. Although fibrinogen and leukocyte count were also positively associated with mortality risk, only CRP remained a significant predictor of mortality when the inflammation markers were adjusted for one another in multivariable models. Serum CRP levels were a long-term predictor of risk of cardiovascular and non-cardiovascular mortality independent of known risk factors, fibrinogen, and leukocyte count.
Collapse
|
248
|
Phillips B, Ranasinghe N, Stewart LA. Ethical and regulatory considerations in the use of individual participant data for studies of disease prediction. Arch Dis Child 2013; 98:567-8. [PMID: 23661573 DOI: 10.1136/archdischild-2013-304149] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
249
|
Stockfelt L, Sallsten G, Almerud P, Basu S, Barregard L. Short-term chamber exposure to low doses of two kinds of wood smoke does not induce systemic inflammation, coagulation or oxidative stress in healthy humans. Inhal Toxicol 2013; 25:417-25. [PMID: 23808634 PMCID: PMC3793281 DOI: 10.3109/08958378.2013.798387] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Introduction: Air pollution increases the risk of cardiovascular diseases. A proposed mechanism is that local airway inflammation leads to systemic inflammation, affecting coagulation and the long-term risk of atherosclerosis. One major source of air pollution is wood burning. Here we investigate whether exposure to two kinds of wood smoke, previously shown to cause airway effects, affects biomarkers of systemic inflammation, coagulation and lipid peroxidation. Methods: Thirteen healthy adults were exposed to filtered air followed by two sessions of wood smoke for three hours, one week apart. One session used smoke from the start-up phase of the wood-burning cycle, and the other smoke from the burn-out phase. Mean particle mass concentrations were 295 µg/m3 and 146 µg/m3, and number concentrations were 140 000/cm3 and 100 000/cm3, respectively. Biomarkers were analyzed in samples of blood and urine taken before and several times after exposure. Results after wood smoke exposure were adjusted for exposure to filtered air. Results: Markers of systemic inflammation and soluble adhesion molecules did not increase after wood smoke exposure. Effects on markers of coagulation were ambiguous, with minor decreases in fibrinogen and platelet counts and mixed results concerning the coagulation factors VII and VIII. Urinary F2-isoprostane, a consistent marker of in vivo lipid peroxidation, unexpectedly decreased after wood smoke exposure. Conclusions: The effects on biomarkers of inflammation, coagulation and lipid peroxidation do not indicate an increased risk of cardiovascular diseases in healthy adults by short-term exposure to wood smoke at these moderate doses, previously shown to cause airway effects.
Collapse
Affiliation(s)
- Leo Stockfelt
- Department of Occupational and Environmental Medicine, Sahlgrenska University Hospital and Academy, Göteborg University, Göteborg, Sweden.
| | | | | | | | | |
Collapse
|
250
|
Abstract
OBJECTIVE To determine whether lower socioeconomic status (SES), broadly defined, is associated with increased inflammation in adolescence and whether adiposity mediates these relationships. METHODS Fasting blood samples from 941 non-Hispanic black and white adolescents enrolled in a suburban, Midwestern school district were assayed for proinflammatory biomarkers (interleukin-6 [IL-6], tumor necrosis factor α soluble receptor 2 fibrinogen). A parent reported objective SES (parent education [E1 ≤ high school, E2 = some college, E3 = college graduate, E4 = professional degree], household income), and youth perceived SES (PSES). Multivariable linear regressions assessed the relationship of SES measures to biomarkers adjusting for age, race, sex, and puberty status. In the final step, body mass index (BMI) z score (BMIz) was added to models, and Sobel tests were performed to assess mediation by adiposity. RESULTS Parent education was inversely associated with IL-6 (βE1 = .11, βE2 = .10, βE3 = .02; p < .001). This association was attenuated but remained significant after BMIz adjustment (p = .01). Sobel testing confirmed BMIz's partial mediating role (p < .001). Parent education was also inversely associated with sTNFR2 (βE1 = .03, βE2 = .02, βE3 = .001; p = .01); this relationship was mediated by BMIz. Although no main effect was noted for PSES, PSES by race interactions was observed for sTNFR2 (p = .02) and IL-6 (p = .06). High PSES was associated with lower sTNFR2 and IL-6 for white but not black youth. There were no associations with household income. CONCLUSIONS Social disadvantage, specifically low parent education, is associated with increased inflammation in adolescence. Adiposity explains some but not all associations, suggesting that other mechanisms link lower SES to inflammation. High PSES is associated with lower inflammation for white but not black youth.
Collapse
|