401
|
The association between high-sensitivity C-reactive protein and metabolic risk factors in black and white South African women: a cross-sectional study. BMC OBESITY 2018; 5:14. [PMID: 29760934 PMCID: PMC5937032 DOI: 10.1186/s40608-018-0191-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 04/22/2018] [Indexed: 11/10/2022]
Abstract
Background High-sensitivity C-reactive protein (hsCRP) is associated with metabolic risk, however it is unclear whether the relationship is confounded by racial/ethnic differences in socioeconomic status (SES), lifestyle factors or central adiposity. The aims of the study was, (1) to investigate whether hsCRP levels differ by race/ethnicity; (2) to examine the race/ethnic-specific associations between hsCRP, HOMA-IR and serum lipids [total cholesterol (TC), triglycerides (TG), high-density lipoproteins (HDL-C) and low-density lipoproteins (LDL-C)]; and (3) to determine whether race/ethnic-specific associations are explained by SES, lifestyle factors or waist circumference (WC). Methods The convenience sample comprised 195 black and 153 white apparently health women, aged 18-45 years. SES (education, assets and housing density) and lifestyle factors (alcohol use, physical activity and contraceptive use) were collected by questionnaire. Weight, height and WC were measured, and fasting blood samples collected for hsCRP, glucose, insulin, and lipids. Results Black women had higher age- and BMI-adjusted hsCRP levels than white women (p = 0.047). hsCRP was associated with HOMA-IR (p < 0.001), TG (p < 0.001), TC (p < 0.05), HDL-C (p < 0.05), and LDL-C (p < 0.05), independent of age and race/ethnicity. The association between hsCRP and lipids differed by race/ethnicity, such that hsCRP was positively associated with TG and LDL-C in white women, and inversely associated with HDL-C in black women. Higher hsCRP was also associated with higher TC in white women and lower TC in black women. Furthermore, when adjusting for SES and lifestyle factors, the associations between hsCRP, and TC and TG, remained, however the associations between hsCRP, and HDL-C and LDL-C, were no longer significant. Conclusion Although circulating hsCRP may identify individuals at increased metabolic risk, the heterogeneity in these associations between racial/ethnic groups highlights the need for prospective studies investigating the role of hsCRP for risk prediction in different populations.
Collapse
|
402
|
Edwards MK, Shivappa N, Mann JR, Hébert JR, Wirth MD, Loprinzi PD. The association between physical activity and dietary inflammatory index on mortality risk in U.S. adults. PHYSICIAN SPORTSMED 2018; 46:249-254. [PMID: 29463180 DOI: 10.1080/00913847.2018.1443665] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Both diet and physical activity have been shown to be associated with reduced risk for mortality. The current study examined the individual and combined effects of the inflammatory quality of diet and physical activity levels on mortality risk. METHODS Data from the 1999-2004 National Health and Nutrition Examination Survey were used (N = 10,610 adults 20-85 years). Participants were followed through 2011, entailing a median follow-up time of 9.6 yrs. Dietary patterns were assessed using the Dietary Inflammatory Index (DII®), with a higher score indicative of greater diet-related inflammation. Habitual physical activity was assessed via a 30-day recall of leisure-time physical activity as well as objectively via up to seven days of accelerometry assessment. Four lifestyle groups were created: 1) unfavorable DII and inactive (referent); 2) unfavorable DII and active; 3) favorable DII and inactive; and 4) favorable DII and active. RESULTS Those who had a low DII score (favorable) and who self-reported being active had a 39% reduced hazard of all-cause mortality (HR = 0.61; 95% CI: 0.49-0.76) when compared to those with a higher DII and were inactive. Results based on accelerometer-assessed physical activity indicate a 65% reduced hazard (HR = 0.35; 95% CI: 0.17-0.74). DII and physical activity also were individually associated with mortality in the expected direction. Specifically, DII was positively associated (HR = 1.08; 95% CI: 1.04-1.13) and physical activity inversely associated (HR = 0.97; 95% CI: 0.94-0.99) with all-cause mortality risk. CONCLUSION The combination of consuming a more anti-inflammatory diet and having adequate levels of physical activity is associated with a reduced risk of mortality. Additional replicative work is warranted.
Collapse
Affiliation(s)
- Meghan K Edwards
- a Physical Activity Epidemiology Laboratory, Exercise Psychology Laboratory, Department of Health, Exercise Science and Recreation Management , University of Mississippi , Oxford , USA
| | - Nitin Shivappa
- b Epidemiology and Biostatistics, The Cancer Prevention and Control Program , University of South Carolina, Connecting Health Innovations, LLC , Columbia , USA
| | - Joshua R Mann
- c Department of Preventive Medicine , University of Mississippi Medical Center, School of Medicine and John D. Bower School of Population Health , Jackson , USA
| | - James R Hébert
- b Epidemiology and Biostatistics, The Cancer Prevention and Control Program , University of South Carolina, Connecting Health Innovations, LLC , Columbia , USA
| | - Michael D Wirth
- b Epidemiology and Biostatistics, The Cancer Prevention and Control Program , University of South Carolina, Connecting Health Innovations, LLC , Columbia , USA
| | - Paul D Loprinzi
- a Physical Activity Epidemiology Laboratory, Exercise Psychology Laboratory, Department of Health, Exercise Science and Recreation Management , University of Mississippi , Oxford , USA
| |
Collapse
|
403
|
Hayek SS, Divers J, Raad M, Xu J, Bowden DW, Tracy M, Reiser J, Freedman BI. Predicting Mortality in African Americans With Type 2 Diabetes Mellitus: Soluble Urokinase Plasminogen Activator Receptor, Coronary Artery Calcium, and High-Sensitivity C-Reactive Protein. J Am Heart Assoc 2018; 7:JAHA.117.008194. [PMID: 29716888 PMCID: PMC6015289 DOI: 10.1161/jaha.117.008194] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Type 2 diabetes mellitus is a major risk factor for cardiovascular disease; however, outcomes in individual patients vary. Soluble urokinase plasminogen activator receptor (suPAR) is a bone marrow-derived signaling molecule associated with adverse cardiovascular and renal outcomes in many populations. We characterized the determinants of suPAR in African Americans with type 2 diabetes mellitus and assessed whether levels were useful for predicting mortality beyond clinical characteristics, coronary artery calcium (CAC), and high-sensitivity C-reactive protein (hs-CRP). METHODS AND RESULTS We measured plasma suPAR levels in 500 African Americans with type 2 diabetes mellitus enrolled in the African American-Diabetes Heart Study. We used Kaplan-Meier curves and Cox proportional hazards models adjusting for clinical characteristics, CAC, and hs-CRP to examine the association between suPAR and all-cause mortality. Last, we report the change in C-statistics comparing the additive values of suPAR, hs-CRP, and CAC to clinical models for prediction of mortality. The suPAR levels were independently associated with female sex, smoking, insulin use, decreased kidney function, albuminuria, and CAC. After a median 6.8-year follow-up, a total of 68 deaths (13.6%) were recorded. In a model incorporating suPAR, CAC, and hs-CRP, only suPAR was significantly associated with mortality (hazard ratio 2.66, 95% confidence interval 1.63-4.34). Addition of suPAR to a baseline clinical model significantly improved the C-statistic for all-cause death (Δ0.05, 95% confidence interval 0.01-0.10), whereas addition of CAC or hs-CRP did not. CONCLUSIONS In African Americans with type 2 diabetes mellitus, suPAR was strongly associated with mortality and improved risk discrimination metrics beyond traditional risk factors, CAC and hs-CRP. Studies addressing the clinical usefulness of measuring suPAR concentrations are warranted.
Collapse
Affiliation(s)
- Salim S Hayek
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Jasmin Divers
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Mohamad Raad
- Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Jianzhao Xu
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC
| | - Donald W Bowden
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC.,Centers for Diabetes Research and Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC
| | - Melissa Tracy
- Department of Medicine, Rush University, Chicago, IL
| | - Jochen Reiser
- Department of Medicine, Rush University, Chicago, IL
| | - Barry I Freedman
- Centers for Diabetes Research and Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC.,Section on Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| |
Collapse
|
404
|
Shimizu H, Hatao F, Imamura K, Takanishi K, Tsujino M. Early Effects of Sleeve Gastrectomy on Obesity-Related Cytokines and Bile Acid Metabolism in Morbidly Obese Japanese Patients. Obes Surg 2018; 27:3223-3229. [PMID: 28569359 DOI: 10.1007/s11695-017-2756-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Laparoscopic sleeve gastrectomy (LSG) has wide-ranging positive effects on adipocytokine metabolism, bile acid profile, and chronic low-grade inflammation related to obesity. However, the early temporal changes in these markers following LSG have not been well investigated. This study aimed to evaluate the early effects of LSG on adipocytokines, bile acid profile, and inflammatory markers. METHODS This was a nonrandomized prospective study examining morbidly obese Japanese patients undergoing LSG. Serial measurements of leptin, adiponectin, bile acids, fibroblast growth factor (FGF)-19, and inflammatory markers were performed preoperatively and 1 and 6 months after LSG. RESULTS The study included ten patients (five females) with a mean age of 48.8 years and BMI 40.9 kg/m2. At baseline, 90% of the patients had T2DM, 70% had dyslipidemia, and 90% had hypertension. Patients lost 5.1 kg/m2 BMI at 1 month and 10.1 kg/m2 BMI at 6 months. The leptin levels sharply decreased, and FGF-19 increased significantly as early as 1 month postoperatively. Adiponectin levels showed an increasing trend at 1 month and a significant increase at 6 months. A significant decrease in high-sensitivity CRP and plasminogen activator inhibitor-1 was observed at 6 months. No significant changes were observed in interleukin (IL)-6, IL-8, tumor necrosis factor-α, serum amyloid A protein, or monocyte chemotactic protein-1 throughout the study. CONCLUSIONS LSG improved the secretion of adipocytokines, increased FGF-19 secretion soon after surgery, and slowly ameliorated inflammation related to obesity through significant weight loss.
Collapse
Affiliation(s)
- Hideharu Shimizu
- Department of Surgery, Tokyo Metropolitan Tama Medical Center, 2-8-29, Musashidai, Fuchu, Tokyo, Japan.
| | - Fumihiko Hatao
- Department of Surgery, Tokyo Metropolitan Tama Medical Center, 2-8-29, Musashidai, Fuchu, Tokyo, Japan
| | - Kazuhiro Imamura
- Department of Surgery, Tokyo Metropolitan Tama Medical Center, 2-8-29, Musashidai, Fuchu, Tokyo, Japan
| | - Kijuro Takanishi
- Department of Surgery, Tokyo Metropolitan Tama Medical Center, 2-8-29, Musashidai, Fuchu, Tokyo, Japan
| | - Motoyoshi Tsujino
- Department of Endocrinology, Tokyo Metropolitan Tama Medical Center, Fuchu, Tokyo, 183-8524, Japan
| |
Collapse
|
405
|
Welsh C, Welsh P, Mark PB, Celis-Morales CA, Lewsey J, Gray SR, Lyall DM, Iliodromiti S, Gill JMR, Pell J, Jhund PS, Sattar N. Association of Total and Differential Leukocyte Counts With Cardiovascular Disease and Mortality in the UK Biobank. Arterioscler Thromb Vasc Biol 2018; 38:1415-1423. [PMID: 29699973 DOI: 10.1161/atvbaha.118.310945] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 04/09/2018] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Elevated white blood cell count is associated with a higher risk of cardiovascular disease (CVD). We aimed to investigate whether specific leukocyte subpopulations, which may more closely indicate a specific inflammatory pathway, are specifically associated with CVD. APPROACH AND RESULTS Participants (478 259) from UK Biobank with data for white blood cell count were included. Death because of CVD (n=1377) and non-CVD causes (n=8987) occurred during median follow-up time of 7.0 years (interquartile range, 6.3-7.6). In Cox models, deciles of leukocyte counts (lymphocytes, monocytes, neutrophils, eosinophils, and basophils) were examined using the fifth decile as the referent group. Models were stratified by sex and adjusted for a range of classical risk factors. A sensitivity analysis excluded participants with baseline comorbidites and the first 2 years of follow-up. Men (hazard ratio [HR], 1.59; 95% confidence interval, 1.22-2.08) and women (HR, 2.15; 95% confidence interval, 1.38-3.35) in the highest decile of neutrophil count were at higher risk of CVD mortality and nonfatal CVD (men HR, 1.28; 95% confidence interval, 1.16-1.42 and women HR, 1.21; 95% confidence interval, 1.06-1.38). In the sensitivity analysis, the power to investigate CVD mortality was limited, but for both sexes combined, the linear HRs for a 1×109/L cell count increase in white blood cell count and neutrophils, respectively, was 1.05 (1.03-1.07) and 1.07 (1.04-1.11). CONCLUSIONS Among circulating leukocyte subpopulations, neutrophil count in men was most consistently associated with fatal and nonfatal CVD. Further studies of interventions that lower circulating neutrophils, such as canakinumab, are required to investigate causality.
Collapse
Affiliation(s)
- Claire Welsh
- From the Institute of Cardiovascular and Medical Sciences (P.W., C.W., P.B.M., C.A.C.-M., S.R.G., S.I., J.M.R.G., P.S.J., N.S.)
| | - Paul Welsh
- From the Institute of Cardiovascular and Medical Sciences (P.W., C.W., P.B.M., C.A.C.-M., S.R.G., S.I., J.M.R.G., P.S.J., N.S.)
| | - Patrick B Mark
- From the Institute of Cardiovascular and Medical Sciences (P.W., C.W., P.B.M., C.A.C.-M., S.R.G., S.I., J.M.R.G., P.S.J., N.S.)
| | - Carlos A Celis-Morales
- From the Institute of Cardiovascular and Medical Sciences (P.W., C.W., P.B.M., C.A.C.-M., S.R.G., S.I., J.M.R.G., P.S.J., N.S.)
| | - James Lewsey
- Institute of Health and Wellbeing (J.L., D.M.L., J.P.), University of Glasgow, Scotland
| | - Stuart R Gray
- From the Institute of Cardiovascular and Medical Sciences (P.W., C.W., P.B.M., C.A.C.-M., S.R.G., S.I., J.M.R.G., P.S.J., N.S.)
| | - Donald M Lyall
- Institute of Health and Wellbeing (J.L., D.M.L., J.P.), University of Glasgow, Scotland
| | - Stamatina Iliodromiti
- From the Institute of Cardiovascular and Medical Sciences (P.W., C.W., P.B.M., C.A.C.-M., S.R.G., S.I., J.M.R.G., P.S.J., N.S.)
| | - Jason M R Gill
- From the Institute of Cardiovascular and Medical Sciences (P.W., C.W., P.B.M., C.A.C.-M., S.R.G., S.I., J.M.R.G., P.S.J., N.S.)
| | - Jill Pell
- Institute of Health and Wellbeing (J.L., D.M.L., J.P.), University of Glasgow, Scotland
| | - Pardeep S Jhund
- From the Institute of Cardiovascular and Medical Sciences (P.W., C.W., P.B.M., C.A.C.-M., S.R.G., S.I., J.M.R.G., P.S.J., N.S.)
| | - Naveed Sattar
- From the Institute of Cardiovascular and Medical Sciences (P.W., C.W., P.B.M., C.A.C.-M., S.R.G., S.I., J.M.R.G., P.S.J., N.S.)
| |
Collapse
|
406
|
Aryan Z, Ghajar A, Faghihi-Kashani S, Afarideh M, Nakhjavani M, Esteghamati A. Baseline High-Sensitivity C-Reactive Protein Predicts Macrovascular and Microvascular Complications of Type 2 Diabetes: A Population-Based Study. ANNALS OF NUTRITION AND METABOLISM 2018; 72:287-295. [PMID: 29694948 DOI: 10.1159/000488537] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 03/16/2018] [Indexed: 01/28/2023]
Abstract
BACKGROUND/AIMS This prospective study is aimed at examining the predictive value of high-sensitivity C-reactive protein (hs-CRP) for coronary heart disease (CHD) events and microvascular complications of type 2 diabetes mellitus (T2DM). METHODS A population-based study (NCT02958579) was conducted on 1,301 participants with T2DM (mean follow-up of 7.5 years). Risk assessment for vascular events was done at baseline, and serum hs-CRP was measured. End points of this study include CHD events, diabetic retinopathy, neuropathy, and diabetic kidney disease. Individuals with unavailable data or hs-CRP >20 mg/L were excluded. The discrimination and reclassification improvement of study end points were tested after addition of hs-CRP to traditional risk factors. RESULTS Median serum hs-CRP was 2.00 ranging from 0.1 to 17 mg/L. Hazards ratio of each SD increment in baseline hs-CRP was 1.028 (1.024-1.032) for CHD, 1.025 (1.021-1.029) for diabetic neuropathy, 1.037 (1.030-1.043) for diabetic retinopathy, and 1.035 (1.027-1.043) for diabetic kidney disease. The addition of hs-CRP to traditional risk factors of vascular complications of T2DM improved discrimination of all end points (p < 0.001). Net reclassification improvement ranged from 8% for diabetic neuropathy to 31% for diabetic kidney disease (p < 0.05). CONCLUSION Baseline hs-CRP predicts both of CHD events and microvascular complications of patients with T2D.
Collapse
Affiliation(s)
- Zahra Aryan
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Student's Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Ghajar
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Faghihi-Kashani
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Afarideh
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Manouchehr Nakhjavani
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Esteghamati
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
407
|
Zhu B, Pan Y, Jing J, Meng X, Zhao X, Liu L, Wang D, Johnston SC, Li H, Wang Y, Wang Z, Wang Y. Neutrophil counts, neutrophil ratio, and new stroke in minor ischemic stroke or TIA. Neurology 2018; 90:e1870-e1878. [DOI: 10.1212/wnl.0000000000005554] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 03/01/2018] [Indexed: 12/20/2022] Open
Abstract
ObjectiveEvidence about whether neutrophil counts or neutrophil ratio is associated with new stroke is scant. The aim of this study is to assess the association of neutrophil counts or neutrophil ratio with a new stroke in patients with minor stroke or TIA.MethodsWe derived data from the Clopidogrel in High-Risk Patients With Acute Nondisabling Cerebrovascular Events trial. Patients with a minor stroke or TIA were categorized into 4 groups according to the quartile of neutrophil counts or neutrophil ratio. The primary outcome was a new stroke (ischemic or hemorrhagic), and secondary outcomes included a new composite vascular event (stroke, myocardial infarction, or death resulting from cardiovascular causes) and ischemic stroke during the 90-day follow-up. We assessed the association between neutrophil counts, neutrophil ratio, and risk of new stroke.ResultsA total of 4,854 participants were enrolled, among whom 495 had new strokes at 90 days. Compared with the first quartile, the second, third, and fourth quartiles of neutrophil counts were associated with increased risk of new stroke (adjusted hazard ratio 1.40 [95% confidence interval (CI) 1.05–1.87], 1.55 [95% CI 1.17–2.05], and 1.69 [95% CI 1.28–2.23], respectively, p for trend <0.001). Similar results were observed for the endpoint of composite events and ischemic stroke. Parallel results were found for neutrophil ratio.ConclusionHigh levels of both neutrophil counts and neutrophil ratio were associated with an increased risk of new stroke, composite events, and ischemic stroke in patients with a minor ischemic stroke or TIA.
Collapse
|
408
|
Sigurdardottir FD, Lyngbakken MN, Holmen OL, Dalen H, Hveem K, Røsjø H, Omland T. Relative Prognostic Value of Cardiac Troponin I and C-Reactive Protein in the General Population (from the Nord-Trøndelag Health [HUNT] Study). Am J Cardiol 2018; 121:949-955. [PMID: 29496193 DOI: 10.1016/j.amjcard.2018.01.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 12/23/2017] [Accepted: 01/04/2018] [Indexed: 01/20/2023]
Abstract
C-reactive protein and cardiac troponin I measured with high-sensitivity assays (high-sensitivity C-reactive protein [hs-CRP] and high-sensitivity troponin I [hs-TnI]) have been associated with risk of fatal and nonfatal cardiovascular events in the general population. The relative prognostic merits of hs-CRP and hs-TnI, and whether these markers of inflammation and subclinical myocardial injury provide incremental information to established cardiovascular risk prediction models, remain unclear. hs-CRP and hs-TnI were measured in 9,005 participants from the prospective observational Nord-Trøndelag Health (HUNT) study. All study subjects were free from known cardiovascular disease at baseline. During a median follow-up period of 13.9 years, 733 participants reached the composite end point of hospitalization for acute myocardial infarction or heart failure, or cardiovascular death. In adjusted models, increased hs-TnI concentrations (>10 ng/L for women and >12 ng/L for men) were associated with the incidence of the composite end point (hazard ratio 3.61, 95% confidence interval [CI] 2.89 to 4.51]), whereas the risk associated with increased hs-CRP concentrations (>3 mg/L for both genders) appeared to be weaker (HR 1.71, 95% CI 1.40 to 2.10). The addition of hs-TnI to established cardiovascular risk prediction models led to a net reclassification improvement of 0.35 (95% CI 0.27 to 0.42), superior to that of hs-CRP (0.21, 95% CI 0.13 to 0.28). The prognostic accuracy of hs-TnI, assessed by C-statistics, was significantly greater than that of hs-CRP (0.753, 95% CI 0.735 to 0.772, vs 0.644, 95% CI 0.625 to 0.663). In conclusion, in subjects from the general population without a history of cardiovascular disease, hs-TnI provides prognostic information superior to that provided by hs-CRP and may therefore be a preferred marker for targeted prevention.
Collapse
Affiliation(s)
- Fjola D Sigurdardottir
- Division of Medicine, Akershus University Hospital, Lørenskog, Norway and Center for Heart Failure Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Magnus N Lyngbakken
- Division of Medicine, Akershus University Hospital, Lørenskog, Norway and Center for Heart Failure Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Oddgeir L Holmen
- HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, Norway; K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Håvard Dalen
- Department of Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Department of Cardiology, St. Olav's University Hospital, Trondheim, Norway
| | - Kristian Hveem
- HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, Norway
| | - Helge Røsjø
- Division of Medicine, Akershus University Hospital, Lørenskog, Norway and Center for Heart Failure Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Torbjørn Omland
- Division of Medicine, Akershus University Hospital, Lørenskog, Norway and Center for Heart Failure Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
409
|
Gouveris H, Bahr K, Jahn C, Matthias C, Simon P. The Apnea-Hypopnea Index Underestimates Systemic Inflammation in Women with Sleep-Disordered Breathing. J Womens Health (Larchmt) 2018; 27:920-926. [PMID: 29630436 DOI: 10.1089/jwh.2017.6819] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Evidence suggests that sleep-related respiratory and related metabolic compromise may vary between females and males with sleep-disordered breathing (SDB). Our purpose was to elucidate possible gender differences in sleep-associated respiratory and inflammatory parameters in patients with SDB. MATERIALS AND METHODS A consecutive number of SDB patients (46 females and 167 males) who underwent polysomnography were retrospectively reviewed. Fibrinogen and C-reactive protein (CRP) concentration, apnea index (ApnI), hypopnea index (HypI), apnea-hypopnea index (AHI), average and minimal SpO2, duration of snoring, age, and body mass index (BMI) were compared between sexes. Spearman's ρ correlation coefficients between parameters were also calculated. RESULTS ApnI and AHI were significantly higher in males. Correlation analysis revealed striking gender differences: only in females, CRP concentration was highly correlated with snoring duration (r = 0.4393), BMI (r = 0.7147), minimal SpO2 (r = -0.4357), and average SpO2 (r = -0.4547); in females, HypI was more strongly correlated with AHI (r = 0.8778), average SpO2 (r = -0.5765), minimal SpO2 (r = -0.5817), and fibrinogen concentration (r = 0.4614) than in males (r = 0.4373; -0.3295; -0.2969; and 0.0887, respectively); in females, age had a much more pronounced effect on ApnI, HypI, AHI, average oxygen saturation (SaO2), minimal SaO2, snoring duration, and CRP and fibrinogen concentration. CONCLUSIONS Respiratory compromise in females with SDB is more strongly associated with systemic inflammation than in males with SDB. Although females display a pathological AHI less frequently than males, they reach quite similar pathological SaO2, CRP, and fibrinogen values. Therefore, AHI may underestimate the pathophysiological systemic effects of SDB in females.
Collapse
Affiliation(s)
- Haralampos Gouveris
- 1 Sleep Medicine Centre and Department of Otorhinolaryngology, Medical Centre of the University of Mainz , Mainz, Germany
| | - Katharina Bahr
- 1 Sleep Medicine Centre and Department of Otorhinolaryngology, Medical Centre of the University of Mainz , Mainz, Germany
| | - Christoph Jahn
- 1 Sleep Medicine Centre and Department of Otorhinolaryngology, Medical Centre of the University of Mainz , Mainz, Germany
| | - Christoph Matthias
- 1 Sleep Medicine Centre and Department of Otorhinolaryngology, Medical Centre of the University of Mainz , Mainz, Germany
| | - Perikles Simon
- 2 Department of Sports Medicine, Rehabilitation and Disease Prevention, University of Mainz , Mainz, Germany
| |
Collapse
|
410
|
Panova-Noeva M, Schulz A, Arnold N, Hermanns MI, Prochaska JH, Laubert-Reh D, Spronk HM, Blettner M, Beutel M, Pfeiffer N, Münzel T, Lackner KJ, Ten Cate H, Wild PS. Coagulation and inflammation in long-term cancer survivors: results from the adult population. J Thromb Haemost 2018; 16:699-708. [PMID: 29431889 DOI: 10.1111/jth.13975] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Indexed: 01/07/2023]
Abstract
Essentials The increase of cancer survival remains curtailed by cardiovascular mortality. We studied a large range of inflammatory and coagulation biomarkers in long-term cancer survivors. Cancer history has an important impact on mortality independent of cardiovascular risk factors. Fibrinogen and von Willebrand factor are potential biomarkers in survivors of increased mortality. SUMMARY Background The advances in cancer treatment and detection of early cancer have resulted in a steady increase in the number of of cancer survivors over the years. However, because of the long-term toxic effects of chemotherapy and radiotherapy, the incidence of cardiovascular disease (CVD) is increasing in survivors. Objectives To investigate traditional cardiovascular risk factors (CVRFs), inflammation and the coagulation profile in long-term cancer survivors (cancer diagnosis ≥ 5 years) from a large adult population-based study sample. Methods The presence of cardiovascular risk factors (CVRFs) and laboratory markers were compared in individuals with (n = 723) and without (n = 13626) a long-term history of cancer from the Gutenberg Health Study. Data on coagulation factors, D-dimer and von Willebrand factor (VWF) activity were available for 4974 individuals (n = 244 cancer survivors). Results In multivariable regression models, a history of cancer was, independently of CVRFs and CVD, associated with higher fibrinogen levels (β 6.99, 95% confidence interval [CI] 1.16-12.8), VWF activity (β 5.08, 95% CI 0.02-10.1), and antithrombin activity (β 1.85, 95% CI 0.44-3.27). Cancer survivors with CVD showed notably higher VWF activity than individuals with CVD without a history of cancer, with a difference in the means of 23.0 (7.9-38.1). Multivariate Cox regression analysis, adjusted for CVRFs, confirmed that a long-term history of cancer is associated with a 72% higher mortality. Increased mortality in cancer survivors was dependent on fibrinogen level and VWF activity level. Conclusion Cancer survivors showed a worse inflammation and coagulation profile than individuals without a history of cancer. Overall mortality in long-term cancer survivors was increased independently of traditional CVRFs. These results underline the need to further investigate plasma biomarkers as complementary cardiovascular risk predictors in cancer survivors.
Collapse
Affiliation(s)
- M Panova-Noeva
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - A Schulz
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - N Arnold
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - M I Hermanns
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - J H Prochaska
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site RhineMain, Mainz, Germany
| | - D Laubert-Reh
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - H M Spronk
- Laboratory for Clinical Thrombosis and Hemostasis, Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, the Netherlands
| | - M Blettner
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - M Beutel
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - N Pfeiffer
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - T Münzel
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site RhineMain, Mainz, Germany
- Center for Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - K J Lackner
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - H Ten Cate
- Laboratory for Clinical Thrombosis and Hemostasis, Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, the Netherlands
| | - P S Wild
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Center for Translational Vascular Biology (CTVB), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site RhineMain, Mainz, Germany
| |
Collapse
|
411
|
Fang M, Qian Q, Zhao Z, Zhu L, Su J, Li X. High-Sensitivity C-Reactive Protein Combined with Low-Density Lipoprotein Cholesterol as the Targets of Statin Therapy in Patients with Acute Coronary Syndrome. Int Heart J 2018; 59:300-306. [PMID: 29479010 DOI: 10.1536/ihj.17-101] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
To investigate the combination of high-sensitivity C-reactive protein (hs-CRP) and Low-density lipoprotein (LDL)-C as the targets for statin treatment in patients with acute coronary syndrome (ACS). This single-center, prospective, randomized study was performed in 400 patients treated with atorvastatin 40 mg/day for 1 month and then with atorvastatin 20 mg/day as maintenance. The patients were randomized to the LDL group (LDL-C target of < 2.07 mmol/L according to the Chinese dyslipidemia guidelines) and to the LDL-CRP group (LDL-C target of < 2.07 mmol/L and hs-CRP target of < 3 mg/L). The patients were followed up for major adverse cardiac events (MACE) at 6, 12, and 18 months. The two groups had similar baseline characteristics and 391 patients completed the follow-up. No differences were found in LDL-C between the two groups, but a difference was found in hs-CRP at 12 and 18 months. There was a significant difference in revascularization (8.7% versus 3.6%, P = 0.04) and MACE (16.8% versus 9.7%; P = 0.04) between the LDL and LDL-CRP groups at 18 months. Compared to LDL-C as the single target, targeting both LDL-C and hs-CRP by statin therapy in patients with ACS could further reduce the incidence of MACE and the residual cardiovascular risk.
Collapse
Affiliation(s)
- Ming Fang
- Department of Cardiology, Hainan General Hospital.,Department of Cardiology, Shanghai Zhoupu Hospital
| | - Qiaohui Qian
- Department of Cardiology, Shanghai Zhoupu Hospital
| | - Zhihong Zhao
- Department of Cardiology, Shanghai Zhoupu Hospital
| | - Luoning Zhu
- Department of Cardiology, Shanghai Zhoupu Hospital
| | - Jinwen Su
- Department of Cardiology, Shanghai Zhoupu Hospital
| | - Xinming Li
- Department of Cardiology, Shanghai Zhoupu Hospital
| |
Collapse
|
412
|
Darweesh SK, Wolters FJ, Ikram MA, Wolf F, Bos D, Hofman A. Inflammatory markers and the risk of dementia and Alzheimer's disease: A meta‐analysis. Alzheimers Dement 2018; 14:1450-1459. [DOI: 10.1016/j.jalz.2018.02.014] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 02/08/2018] [Accepted: 02/16/2018] [Indexed: 11/16/2022]
Affiliation(s)
- Sirwan K.L. Darweesh
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMAUSA
- Department of EpidemiologyErasmus MC University Medical Center RotterdamRotterdamthe Netherlands
- Department of NeurologyErasmus MC University Medical Center RotterdamRotterdamthe Netherlands
| | - Frank J. Wolters
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMAUSA
- Department of EpidemiologyErasmus MC University Medical Center RotterdamRotterdamthe Netherlands
- Department of NeurologyErasmus MC University Medical Center RotterdamRotterdamthe Netherlands
| | - M. Arfan Ikram
- Department of EpidemiologyErasmus MC University Medical Center RotterdamRotterdamthe Netherlands
- Department of NeurologyErasmus MC University Medical Center RotterdamRotterdamthe Netherlands
- Department of Radiology and Nuclear MedicineErasmus MC University Medical Center RotterdamRotterdamthe Netherlands
| | - Frank Wolf
- Janssen Prevention CenterLeidenthe Netherlands
- Faculty of MedicineSchool of Public HealthImperial College LondonLondonUK
| | - Daniel Bos
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMAUSA
- Department of EpidemiologyErasmus MC University Medical Center RotterdamRotterdamthe Netherlands
- Department of Radiology and Nuclear MedicineErasmus MC University Medical Center RotterdamRotterdamthe Netherlands
| | - Albert Hofman
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMAUSA
- Department of EpidemiologyErasmus MC University Medical Center RotterdamRotterdamthe Netherlands
| |
Collapse
|
413
|
Thunström E, Glantz H, Yucel-Lindberg T, Lindberg K, Saygin M, Peker Y. CPAP Does Not Reduce Inflammatory Biomarkers in Patients With Coronary Artery Disease and Nonsleepy Obstructive Sleep Apnea: A Randomized Controlled Trial. Sleep 2018; 40:4345662. [PMID: 29029237 DOI: 10.1093/sleep/zsx157] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objectives Obstructive sleep apnea (OSA) and enhanced vascular inflammation coexist in patients with coronary artery disease (CAD). Continuous positive airway pressure (CPAP) is first-line treatment for OSA with daytime sleepiness. This analysis of data from the RICCADSA (Randomized Intervention with CPAP in Coronary Artery Disease and Sleep Apnea) trial investigated the effects of CPAP on inflammatory markers in patients with CAD and nonsleepy OSA. Methods This single-center, randomized, controlled, open-label trial enrolled consecutive revascularized patients with nonsleepy OSA (apnea-hypopnea index >15/h; Epworth Sleepiness Scale score <10). Levels of high-sensitivity C-reactive protein (hs-CRP), interleukin (IL)-6, IL-8, and tumor necrosis factor-α (TNF-α) were measured in blood samples taken at baseline (median 94 days after revascularization) and after 1 year of follow-up in patients randomized to CPAP or no-CPAP. Results A total of 220 patients with analyzable blood samples at baseline and 1 year were included. Baseline IL-6 levels were significantly lower in the CPAP versus no-CPAP group (median 3.1 pmol/L [interquartile range 1.3-5.7] vs. 4.2 pmol/L [2.0-8.9], respectively; p = .005). At 1-year follow-up, median IL-6 levels were significantly reduced in both groups (to 2.2 pmol/L [1.2-3.9] in the CPAP group and to 2.2 [1.2-4.7] in no-CPAP group; both p < .001 vs. baseline). IL-8, hs-CRP, and TNF-α did not change significantly from baseline. There was no association between CPAP adherence and changes in inflammatory marker levels. Conclusions In patients with stable CAD and nonsleepy OSA, inflammatory biomarkers did not change significantly over time except for IL-6 levels, which reduced to the same extent in the CPAP and no-CPAP groups. Clinical Trial Registration ClinicalTrials.gov, ID: NCT00519597; researchweb.org, VGSKAS-4731.
Collapse
Affiliation(s)
- Erik Thunström
- Department of Molecular and Clinical Medicine, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Helena Glantz
- Department of Molecular and Clinical Medicine, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Internal Medicine, Skaraborg Hospital, Lidköping, Sweden
| | - Tülay Yucel-Lindberg
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Kristin Lindberg
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Mustafa Saygin
- Department of Physiology, Faculty of Medicine, Süleyman Demirel University, Isparta, Turkey
| | - Yüksel Peker
- Department of Molecular and Clinical Medicine, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Pulmonary Medicine, Marmara University, Istanbul, Turkey
| |
Collapse
|
414
|
Novel and Emerging Biomarkers with Risk Predictive Utility for Atherosclerotic Cardiovascular Disease. CURRENT CARDIOVASCULAR RISK REPORTS 2018. [DOI: 10.1007/s12170-018-0570-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
415
|
Badimon L, Peña E, Arderiu G, Padró T, Slevin M, Vilahur G, Chiva-Blanch G. C-Reactive Protein in Atherothrombosis and Angiogenesis. Front Immunol 2018; 9:430. [PMID: 29552019 PMCID: PMC5840191 DOI: 10.3389/fimmu.2018.00430] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/16/2018] [Indexed: 12/11/2022] Open
Abstract
C-reactive protein (CRP) is a short pentraxin mainly found as a pentamer in the circulation, or as non-soluble monomers CRP (mCRP) in tissues, exerting different functions. This review is focused on discussing the role of CRP in cardiovascular disease, including recent advances on the implication of CRP and its forms specifically on the pathogenesis of atherothrombosis and angiogenesis. Besides its role in the humoral innate immune response, CRP contributes to cardiovascular disease progression by recognizing and binding multiple intrinsic ligands. mCRP is not present in the healthy vessel wall but it becomes detectable in the early stages of atherogenesis and accumulates during the progression of atherosclerosis. CRP inhibits endothelial nitric oxide production and contributes to plaque instability by increasing endothelial cell adhesion molecules expression, by promoting monocyte recruitment into the atheromatous plaque and by enzymatically binding to modified low-density lipoprotein. CRP also contributes to thrombosis, but depending on its form it elicits different actions. Pentameric CRP has no involvement in thrombogenesis, whereas mCRP induces platelet activation and thrombus growth. In addition, mCRP has apparently contradictory pro-angiogenic and anti-angiogenic effects determining tissue remodeling in the atherosclerotic plaque and in infarcted tissues. Overall, CRP contributes to cardiovascular disease by several mechanisms that deserve an in-depth analysis.
Collapse
Affiliation(s)
- Lina Badimon
- Cardiovascular Science Institute - ICCC, IIB-Sant Pau, Hospital de Sant Pau, Barcelona, Spain.,CiberCV, Institute Carlos III, Madrid, Spain
| | - Esther Peña
- Cardiovascular Science Institute - ICCC, IIB-Sant Pau, Hospital de Sant Pau, Barcelona, Spain.,CiberCV, Institute Carlos III, Madrid, Spain
| | - Gemma Arderiu
- Cardiovascular Science Institute - ICCC, IIB-Sant Pau, Hospital de Sant Pau, Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Science Institute - ICCC, IIB-Sant Pau, Hospital de Sant Pau, Barcelona, Spain.,CiberCV, Institute Carlos III, Madrid, Spain
| | - Mark Slevin
- School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
| | - Gemma Vilahur
- Cardiovascular Science Institute - ICCC, IIB-Sant Pau, Hospital de Sant Pau, Barcelona, Spain.,CiberCV, Institute Carlos III, Madrid, Spain
| | - Gemma Chiva-Blanch
- Cardiovascular Science Institute - ICCC, IIB-Sant Pau, Hospital de Sant Pau, Barcelona, Spain
| |
Collapse
|
416
|
Hwang Y, Morrow DA, Cannon CP, Liu Y, Bergenstal R, Heller S, Mehta C, Cushman W, Bakris GL, Zannad F, White WB. High-sensitivity C-reactive protein, low-density lipoprotein cholesterol and cardiovascular outcomes in patients with type 2 diabetes in the EXAMINE (Examination of Cardiovascular Outcomes with Alogliptin versus Standard of Care) trial. Diabetes Obes Metab 2018; 20:654-659. [PMID: 29064626 PMCID: PMC5836896 DOI: 10.1111/dom.13136] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 09/28/2017] [Accepted: 10/19/2017] [Indexed: 12/28/2022]
Abstract
AIMS We sought to assess the risk of major adverse cardiovascular events (MACE) by utilizing high-sensitivity C-reactive protein (hsCRP) level and low-density lipoprotein cholesterol (LDL-C) in patients with type 2 diabetes and recent acute coronary syndrome. MATERIALS AND METHODS Study participants enrolled in the EXAMINE trial (Clinical trials registration number: NCT00968708) and were stratified by baseline hsCRP levels (<1, 1-3 and >3 mg/L). They were also sub-divided into 4 groups according to baseline hsCRP (≤3 or >3 mg/L) and achieved LDL-C (<70 or ≥70 mg/dL) levels. Among 5380 patients, the MACE rate, a composite of cardiovascular death, non-fatal acute myocardial infarction and non-fatal stroke, was evaluated during the 30 months of follow-up. RESULTS Cumulative incidence of MACE was 11.5% (119 events), 14.6% (209 events) and 18.4% (287 events) in patients with hsCRP levels of <1, 1 to 3 and >3 mg/L, respectively (P < .001). In patients with hsCRP >3 mg/L, the adjusted hazard ratio (95% confidence interval) was 1.42 (1.13, 1.78; P = .002) for MACE compared with patients with hsCRP <1 mg/L. MACE cumulative incidences were 11.0% (128 events), 14.4% (100 events), 15.6% (194 events) and 21.3% (182 events) in patients with low LDL-C and low hsCRP, low LDL-C and high hsCRP, high LDL-C and low hsCRP, and high LDL-C and high hsCRP levels, respectively (P < .001). CONCLUSIONS Levels of hsCRP were associated with recurrent cardiovascular events in patients with type 2 diabetes and recent acute coronary syndrome, and this association appears to be independent of and additive to the achieved LDL-C level.
Collapse
Affiliation(s)
- You‐Cheol Hwang
- Division of Endocrinology and Metabolism, Department of MedicineKyung Hee University School of Medicine, Kyung Hee University Hospital at GangdongSeoulSouth Korea
| | - David A. Morrow
- Cardiovascular DivisionBrigham and Women's HospitalBostonMassachusetts
| | | | - Yuyin Liu
- Baim Institute for Clinical ResearchBostonMassachusetts
| | | | | | - Cyrus Mehta
- Harvard School of Public HealthBostonMassachusetts
| | | | | | | | - William B. White
- University of Connecticut School of MedicineFarmingtonConnecticut
| |
Collapse
|
417
|
Dalsgaard NB, Vilsbøll T, Knop FK. Effects of glucagon-like peptide-1 receptor agonists on cardiovascular risk factors: A narrative review of head-to-head comparisons. Diabetes Obes Metab 2018; 20:508-519. [PMID: 29024408 PMCID: PMC5836903 DOI: 10.1111/dom.13128] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/15/2017] [Accepted: 10/09/2017] [Indexed: 12/11/2022]
Abstract
Cardiovascular (CV) disease is the leading cause of death and morbidity in patients with type 2 diabetes. Five CV risk factors (blood pressure, resting heart rate, body weight, cholesterol levels and blood glucose) are monitored routinely as safety and efficacy endpoints in randomized clinical trials for diabetes therapies. To determine if different glucagon-like peptide-1 receptor agonists (GLP-1RAs) had varying effects on these CV risk factors, we reviewed 16 head-to-head trials directly comparing GLP-1RAs that included at least one of the five factors. Few trials reported statistical differences between GLP-1RAs in terms of systolic blood pressure (SBP), body weight and total cholesterol. Liraglutide increased heart rate vs its comparators in three separate trials. All GLP-1RAs reduced glycated haemoglobin (HbA1c), but exenatide twice daily and lixisenatide had statistically smaller effects compared with other GLP-1RAs. These descriptive data indicate that individual GLP-1RAs affect CV risk factors differently, potentially because of their individual pharmacokinetics and/or size. Short-acting GLP-1RAs appeared to result in smaller changes in SBP and total cholesterol compared with continuous-acting treatments, while large GLP-1RAs had a reduced effect on body weight compared with small GLP-1RAs. For glycaemic control, short-acting GLP-1RAs had a greater impact on postprandial glucose levels vs continuous-acting GLP-1RAs, but for fasting plasma glucose levels and HbA1c, continuous-acting treatments had the greater effect. No differentiating trends were obvious in heart rate data. These diverse actions of GLP-1RAs on CV risk factors should aid individualized patient treatment.
Collapse
Affiliation(s)
- Niels B. Dalsgaard
- Center for Diabetes Research, Gentofte Hospital, University of CopenhagenHellerupDenmark
| | - Tina Vilsbøll
- Center for Diabetes Research, Gentofte Hospital, University of CopenhagenHellerupDenmark
- Steno Diabetes Center Copenhagen, University of CopenhagenGentofteDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Filip K. Knop
- Center for Diabetes Research, Gentofte Hospital, University of CopenhagenHellerupDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
418
|
Lai CL, Xing JP, Liu XH, Qi J, Zhao JQ, Ji YR, Yang WX, Yan PJ, Luo CY, Ruan LF. Relationships of Inflammatory Factors and Risk Factors with Different Target Organ Damage in Essential Hypertension Patients. Chin Med J (Engl) 2018; 130:1296-1302. [PMID: 28524828 PMCID: PMC5455038 DOI: 10.4103/0366-6999.206343] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Atherosclerosis (AS) is an inflammatory disease. Inflammation was considered to play a role in the whole process of AS. This study aimed to analyze the relationships of inflammatory factors and risk factors with different target organ damages (TOD) in essential hypertension (EH) patients and to explore its clinical significance. METHODS A total of 294 EH patients were selected and divided into four groups according to their conditions of TOD. Forty-eight healthy subjects were selected as control. The clinical biochemical parameters, serum amyloid A, serum tryptase, and lipoprotein-associated phospholipase A2 (Lp-PLA2) in each group were detected, and the related risk factors were also statistically analyzed. RESULTS Fibrinogen (Fbg) was the most significant independent risk factor in acute coronary syndrome (ACS) group (odds ratio [OR]: 22.242, 95% confidence interval [CI]: 6.458-76.609, P< 0.001) with the largest absolute value of the standardized partial regression coefficient B' (b': 1.079). Lp-PLA2 was the most significant independent risk factor in stroke group (OR: 13.699, 95% CI: 5.236-35.837, P< 0.001) with b' = 0.708. Uric acid (UA) was the most significant independent risk factor in renal damage group (OR: 15.307, 95% CI: 4.022-58.250, P< 0.001) with b' = 1.026. CONCLUSIONS Fbg, Lp-PLA2, and UA are the strongest independent risk factors toward the occurrence of ACS, ischemic stroke, and renal damage in EH patients, thus exhibiting the greatest impacts on the occurrence of ACS, ischemic stroke, and renal damage in EH patients, respectively.
Collapse
Affiliation(s)
- Chun-Lin Lai
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Jin-Ping Xing
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Xiao-Hong Liu
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Jie Qi
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Jian-Qiang Zhao
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - You-Rui Ji
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Wu-Xiao Yang
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Pu-Juan Yan
- Postgraduate School of Shanxi Medical University, Taiyuan, Shanxi 030012, China
| | - Chun-Yan Luo
- Postgraduate School of Shanxi Medical University, Taiyuan, Shanxi 030012, China
| | - Lu-Fang Ruan
- Postgraduate School of Shanxi Medical University, Taiyuan, Shanxi 030012, China
| |
Collapse
|
419
|
Bocchia M, Galimberti S, Aprile L, Sicuranza A, Gozzini A, Santilli F, Abruzzese E, Baratè C, Scappini B, Fontanelli G, Trawinska MM, Defina M, Gozzetti A, Bosi A, Petrini M, Puccetti L. Genetic predisposition and induced pro-inflammatory/pro-oxidative status may play a role in increased atherothrombotic events in nilotinib treated chronic myeloid leukemia patients. Oncotarget 2018; 7:72311-72321. [PMID: 27527867 PMCID: PMC5342164 DOI: 10.18632/oncotarget.11100] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/1969] [Accepted: 07/18/2016] [Indexed: 12/18/2022] Open
Abstract
Several reports described an increased risk of cardiovascular (CV) events, mainly atherothrombotic, in Chronic Myeloid Leukemia (CML) patients receiving nilotinib. However, the underlying mechanism remains elusive. The objective of the current cross-sectional retrospective study is to address a potential correlation between Tyrosine Kinase Inhibitors (TKIs) treatment and CV events. One hundred and 10 chronic phase CML patients in complete cytogenetic response during nilotinib or imatinib, were screened for CV events and evaluated for: traditional CV risk factors, pro/anti-inflammatory biochemical parameters and detrimental ORL1 gene polymorphisms (encoding for altered oxidized LDL receptor-1). Multivariate analysis of the whole cohort showed that the cluster of co-existing nilotinib treatment, dyslipidaemia and G allele of LOX-1 polymorphism was the only significant finding associated with CV events. Furthermore, multivariate analysis according to TKI treatment confirmed IVS4-14 G/G LOX-1 polymorphism as the strongest predictive factor for a higher incidence of CV events in nilotinib patients. Biochemical assessment showed an unbalanced pro-inflammatory cytokines network in nilotinib vs imatinib patients. Surprisingly, pre-existing traditional CV risk factors were not always predictive of CV events. We believe that in nilotinib patients an induced “inflammatory/oxidative status”, together with a genetic pro-atherothrombotic predisposition, may favour the increased incidence of CV events. Prospective studies focused on this issue are ongoing.
Collapse
Affiliation(s)
- Monica Bocchia
- Department of Hematology, University of Siena, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Sara Galimberti
- Department of Clinical and Experimental Medicine, U.O. Hematology, University of Pisa, Pisa, Italy
| | - Lara Aprile
- Department of Hematology, University of Siena, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Anna Sicuranza
- Department of Hematology, University of Siena, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Antonella Gozzini
- Functional Unit of Hematology, University of Florence, Florence, Italy
| | | | | | - Claudia Baratè
- Department of Clinical and Experimental Medicine, U.O. Hematology, University of Pisa, Pisa, Italy
| | - Barbara Scappini
- Functional Unit of Hematology, University of Florence, Florence, Italy
| | - Giulia Fontanelli
- Department of Clinical and Experimental Medicine, U.O. Hematology, University of Pisa, Pisa, Italy
| | | | - Marzia Defina
- Department of Hematology, University of Siena, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Alessandro Gozzetti
- Department of Hematology, University of Siena, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Alberto Bosi
- Functional Unit of Hematology, University of Florence, Florence, Italy
| | - Mario Petrini
- Department of Clinical and Experimental Medicine, U.O. Hematology, University of Pisa, Pisa, Italy
| | - Luca Puccetti
- Department of Hematology, University of Siena, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| |
Collapse
|
420
|
Lassale C, Curtis A, Abete I, van der Schouw YT, Verschuren WMM, Lu Y, Bueno-de-Mesquita HBA. Elements of the complete blood count associated with cardiovascular disease incidence: Findings from the EPIC-NL cohort study. Sci Rep 2018; 8:3290. [PMID: 29459661 PMCID: PMC5818488 DOI: 10.1038/s41598-018-21661-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 02/08/2018] [Indexed: 12/31/2022] Open
Abstract
All blood cells (white blood cells [WBC], red blood cells [RBC] and platelets) can play a role in atherosclerosis. Complete blood count (CBC) is widely available in clinical practice but utility as potential risk factors for cardiovascular disease (CVD) is uncertain. Our aim was to assess the associations of pre-diagnostic CBC with incidence of CVD in 14,362 adults free of CVD and aged 47.8 (±11.7) years at baseline, followed-up for 11.4 years (992 incident cases). Cox proportional hazards regressions were used to estimate HRs and 95%CI. Comparing the top (T3) to bottom (T1) tertile, increased total WBC, lymphocyte, monocyte and neutrophil counts were associated with higher CVD risk: 1.31 (1.10; 1.55), 1.20 (1.02; 1.41), 1.21 (1.03; 1.41) and 1.24 (1.05; 1.47), as well as mean corpuscular volume (MCV: 1.23 [1.04; 1.46]) and red cell distribution width (RDW: 1.22 [1.03; 1.44]). Platelets displayed an association for count values above the clinically normal range: 1.49 (1.00; 2.22). To conclude, total and differential WBC count, MCV, RDW and platelet count likely play a role in the aetiology of CVD but only WBC provide a modest improvement for the prediction of 10-year CVD risk over traditional CVD risk factors in a general population.
Collapse
Affiliation(s)
- Camille Lassale
- Department of Epidemiology and Public Health, University College London, 1-19 Torrington Place, London, WC1E 7HB, United Kingdom.
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, Norfolk Place, London, W2 1PG, United Kingdom.
| | - Alyscia Curtis
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, Norfolk Place, London, W2 1PG, United Kingdom
| | - Itziar Abete
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, Norfolk Place, London, W2 1PG, United Kingdom
- Nutrition Research Center, University of Navarra, 31010, Pamplona, Spain
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - W M Monique Verschuren
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
- Center for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment (RIVM), 3720 BA, Bilthoven, The Netherlands
| | - Yunxia Lu
- Program in Public Health, College of Health Sciences, University of California Irvine, Irvine, CA, United States of America
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - H B As Bueno-de-Mesquita
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, Norfolk Place, London, W2 1PG, United Kingdom
- Center for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment (RIVM), 3720 BA, Bilthoven, The Netherlands
- Department of Social & Preventive Medicine, Faculty of Medicine, University of Malaya, Pantai Valley, 50603, Kuala Lumpur, Malaysia
| |
Collapse
|
421
|
Subirana I, Fitó M, Diaz O, Vila J, Francés A, Delpon E, Sanchis J, Elosua R, Muñoz-Aguayo D, Dégano IR, Marrugat J. Prediction of coronary disease incidence by biomarkers of inflammation, oxidation, and metabolism. Sci Rep 2018; 8:3191. [PMID: 29453342 PMCID: PMC5816603 DOI: 10.1038/s41598-018-21482-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/31/2018] [Indexed: 01/16/2023] Open
Abstract
The effect of circulating biomarkers in predicting coronary artery disease (CAD) is not fully elucidated. This study aimed to determine the relationship with CAD and the predictive capacity of nine biomarkers of inflammation (TNF-α, IL-10, IL-6, MCP-1, CRP), oxidation (GHS-Px), and metabolism (adiponectin, leptin, and insulin). This was a case-cohort study, within the REGICOR population-cohorts (North-Eastern Spain), of 105 CAD cases and 638 individuals randomly selected from a cohort of 5,404 participants aged 35–74 years (mean follow-up = 6.1 years). Biomarkers’ hazard ratio (HR)/standard deviation was estimated with Cox models adjusted for age, sex, and classical risk factors. Discrimination improvement and reclassification were analyzed with the c-index and the Net reclassification index (NRI). GHS-Px (adjusted HRs = 0.77; 95%CI:0.60–0.99), insulin (1.46; 1.08–1.98), leptin (1.40; 1.03–1.90), IL-6 (1.34; 1.03–1.74), and TNF-α (1.80; 1.26–2.57) were significantly associated with CAD incidence. In the model adjusted for all biomarkers, TNF-α (1.87;1.31–2.66) and insulin (1.59;1.16–2.19) were independently associated with CAD. This final model, compared to a model without biomarkers, showed a c-index difference of 1.3% (−0.7, 3.2) and a continuous NRI of 33.7% (2.6, 61.9). TNF-α and insulin are independently associated with CAD incidence and they improve reclassification when added to a model including classical risk factors.
Collapse
Affiliation(s)
- Isaac Subirana
- CIBER of Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Cardiovascular Epidemiology and Genetics Research Group, Program of Epidemiology and Public Health, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Montserrat Fitó
- Cardiovascular Risk and Nutrition Research Group, Program of Epidemiology and Public Health, IMIM, Barcelona, Spain.,CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), ISCIII, Madrid, Spain
| | - Oscar Diaz
- Cardiovascular Epidemiology and Genetics Research Group, Program of Epidemiology and Public Health, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Joan Vila
- CIBER of Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Cardiovascular Epidemiology and Genetics Research Group, Program of Epidemiology and Public Health, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | | | | | - Juan Sanchis
- Servei de Cardiologia, INCLIVA, Departament de Medicina, Hospital Clínico Universitario de Valencia, Universitat de Valencia, Valencia, Spain
| | - Roberto Elosua
- Cardiovascular Epidemiology and Genetics Research Group, Program of Epidemiology and Public Health, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.,CIBER of Cardiovascular Diseases (CIBERCV), ISCIII, Madrid, Spain
| | - Daniel Muñoz-Aguayo
- Cardiovascular Risk and Nutrition Research Group, Program of Epidemiology and Public Health, IMIM, Barcelona, Spain.,CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), ISCIII, Madrid, Spain
| | - Irene R Dégano
- CIBER of Cardiovascular Diseases (CIBERCV), ISCIII, Madrid, Spain. .,REGICOR Group, Program of Epidemiology and Public Health, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.
| | - Jaume Marrugat
- CIBER of Cardiovascular Diseases (CIBERCV), ISCIII, Madrid, Spain. .,REGICOR Group, Program of Epidemiology and Public Health, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.
| |
Collapse
|
422
|
Correlation between global methylation level of peripheral blood leukocytes and serum C reactive protein level modified by MTHFR polymorphism: a cross-sectional study. BMC Cancer 2018; 18:184. [PMID: 29439678 PMCID: PMC5812223 DOI: 10.1186/s12885-018-4089-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 02/05/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Chronic inflammatory conditions are associated with higher tumor incidence through epigenetic and genetic alterations. Here, we focused on an association between an inflammation marker, C-reactive-protein (CRP), and global DNA methylation levels of peripheral blood leukocytes. METHODS The subjects were 384 healthy Japanese women enrolled as the control group of a case-control study for breast cancer conducted from 2001 to 2005. Global DNA methylation was quantified by Luminometric Methylation Assay (LUMA). RESULTS With adjustment for lifestyle-related factors, including folate intake, the global DNA methylation level of peripheral blood leukocytes was significantly but weakly increased by 0.43% per quartile category for CRP (P for trend = 0.010). Estimated methylation levels stratified by CRP quartile were 70.0%, 70.8%, 71.4%, and 71.3%, respectively. In addition, interaction between polymorphism of MTHFR (rs1801133, known as C677T) and CRP was significant (P for interaction = 0.046); the global methylation level was significantly increased by 0.61% per quartile category for CRP in the CT/TT group (those with the minor allele T, P for trend = 0.001), whereas no association was observed in the CC group (wild type). CONCLUSIONS Our study suggests that CRP concentration is weakly associated with global DNA methylation level. However, this association was observed more clearly in individuals with the minor allele of the MTHFR missense SNP rs1801133. By elucidating the complex mechanism of the regulation of DNA methylation by both acquired and genetic factors, our results may be important for cancer prevention.
Collapse
|
423
|
Kelly PJ, Murphy S, Coveney S, Purroy F, Lemmens R, Tsivgoulis G, Price C. Anti-inflammatory approaches to ischaemic stroke prevention. J Neurol Neurosurg Psychiatry 2018; 89:211-218. [PMID: 28935831 DOI: 10.1136/jnnp-2016-314817] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 07/24/2017] [Accepted: 08/08/2017] [Indexed: 12/19/2022]
Abstract
Stroke is a major cause of neurological morbidity and mortality. Atherosclerosis is a major contributor to first and recurrent stroke. A growing evidence base indicates that inflammation is a key process in the pathogenesis of atherosclerosis, leading to thromboembolic events. In this review, we summarise the evidence linking inflammation to stroke risk and discuss clinical trials addressing the 'inflammation hypothesis' in coronary disease and stroke. Trial registration number CONVINCE trial ClinicalTrials.gov number; NCT 02898610; Pre-results.
Collapse
Affiliation(s)
- Peter J Kelly
- Health Research Board Irish Stroke Clinical Trials Network and Mater University Hospital/University College Dublin, Dublin, Ireland
| | - Sean Murphy
- Royal College of Surgeons Ireland, Dublin, Ireland.,Mater University Hospital/University College Dublin, Dublin, Ireland
| | - Sarah Coveney
- Health Research Board Irish Stroke Clinical Trials Network and Mater University Hospital/University College Dublin, Dublin, Ireland
| | - Francisco Purroy
- Stroke Unit, Department of Neurology, Hospitalt Universitari Arnau de Vilanova de Lleida and Universitat de Lleida, Biomedical Research Institute of Lleida, Universitat de Lleida, Spain, Dublin, Ireland
| | - Robin Lemmens
- Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND), KU Leuven - University of Leuven, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium.,VIB Center for Brain and Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Georgios Tsivgoulis
- Second Department of Neurology, "Attikon" Hospital, University of Athens, School of Medicine, Athens, Greece.,Department of Neurology, University of Tennessee Health Science Center, Memphis, TN
| | - Chris Price
- Institute of Neuroscience, Newcastle University, Newcastle, UK
| |
Collapse
|
424
|
Maestro de la Calle G, Fernández-Ruiz M, López-Medrano F, Polanco N, González E, San Juan R, Ruiz-Merlo T, Origüen J, Paz-Artal E, Andrés A, Aguado JM. Post-transplant hypocomplementemia: A novel marker of cardiovascular risk in kidney transplant recipients? Atherosclerosis 2018; 269:204-210. [PMID: 29407595 DOI: 10.1016/j.atherosclerosis.2018.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 12/22/2017] [Accepted: 01/12/2018] [Indexed: 10/18/2022]
Abstract
BACKGROUND AND AIMS Cardiovascular disease (CVD) is a leading cause of mortality after kidney transplantation (KT). The potential role of the complement system in the pathogenesis of post-transplant CVD remains unexplored. METHODS Serum complement (C3 and C4) levels were measured at baseline and post-transplant months 1 and 6 in 447 kT recipients. The study outcome was post-transplant atherothrombotic event (PAE), a composite of acute coronary syndrome, critical peripheral arterial disease, stroke and/or transient ischemic attack. RESULTS After a median follow-up of 4.2 years, 48 PAEs occurred in 43 patients (cumulative incidence: 9.6%; incidence rate: 2.6 events per 100 transplant-years). No differences were found in C3 and C4 levels at baseline or month 1 between patients with or without PAE. However, C3 levels at month 6 were significantly lower in patients developing PAE beyond that point (i.e., late PAE) (96.9 ± 22.3 vs. 109.6 ± 24.0 mg/dL; p = 0.013). The presence of C3 hypocomplementemia at month 6 was associated with a lower PAE-free survival (p = 0.002). After adjusting for conventional CVD risk factors and acute graft rejection, C3 hypocomplementemia at month 6 remained as an independent risk factor for late PAE in all the exploratory models (minimum hazard ratio: 3.24; p = 0.011). With respect to a model exclusively based on clinical variables, the inclusion of C3 levels at month 6 improved predictive capacity (areas under ROC curves: 0.788 and 0.812, respectively). CONCLUSIONS Post-transplant monitoring of serum C3 levels might be useful to identify KT recipients at increased risk of CVD.
Collapse
Affiliation(s)
- Guillermo Maestro de la Calle
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Mario Fernández-Ruiz
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain.
| | - Francisco López-Medrano
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Natalia Polanco
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Esther González
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Rafael San Juan
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Tamara Ruiz-Merlo
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Julia Origüen
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Estela Paz-Artal
- Department of Immunology, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Amado Andrés
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - José María Aguado
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| |
Collapse
|
425
|
Otvos JD, Guyton JR, Connelly MA, Akapame S, Bittner V, Kopecky SL, Lacy M, Marcovina SM, Muhlestein JB, Boden WE. Relations of GlycA and lipoprotein particle subspecies with cardiovascular events and mortality: A post hoc analysis of the AIM-HIGH trial. J Clin Lipidol 2018; 12:348-355.e2. [PMID: 29409728 DOI: 10.1016/j.jacl.2018.01.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/28/2017] [Accepted: 01/03/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND The Atherothrombosis Intervention in Metabolic Syndrome with Low HDL/High Triglycerides and Impact on Global Health Outcomes trial showed no incremental benefit of extended-release niacin (ERN) therapy added to simvastatin in subjects with cardiovascular disease (CVD). OBJECTIVES To examine the effects of ERN treatment on lipoprotein particles and GlycA, a new marker of systemic inflammation, and their relations with incident CVD events including mortality. METHODS GlycA and very low-density lipoprotein, low-density lipoprotein (LDL), and high-density lipoprotein (HDL) particle subclasses were quantified by nuclear magnetic resonance spectroscopy using available stored baseline (n = 2754) and 1-year in-trial (n = 2581) samples. Associations with CVD events and all-cause mortality were assessed using multivariable Cox proportional hazards regression adjusted for age, sex, diabetes, treatment assignment, and lipoproteins. RESULTS Compared to placebo, ERN treatment lowered very low-density lipoprotein and LDL and increased HDL particle concentrations, increased LDL and HDL particle sizes (all P < .0001), but did not affect GlycA. Baseline and in-trial GlycA levels were associated with increased risk of CVD events: hazard ratio (HR) per SD increment, 1.17 (95% confidence interval [CI], 1.06-1.28) and 1.13 (1.02-1.26), respectively. However, none of the lipoprotein particle classes or subclasses was associated with incident CVD. By contrast, all-cause mortality was significantly associated with both GlycA (baseline HR: 1.46 [1.22-1.75]; in-trial HR: 1.41 [1.24-1.60]) and low levels of small HDL particles (baseline HR: 0.69 [0.56-0.86]; in-trial HR: 0.69 [0.56-0.86]). CONCLUSIONS This Atherothrombosis Intervention in Metabolic Syndrome with Low HDL/High Triglycerides and Impact on Global Health Outcomes trial post hoc substudy indicates that inflammation, as indexed by GlycA, is unaffected by ERN treatment but is significantly associated with the residual risk of CVD and death in patients treated to low levels of LDL cholesterol.
Collapse
Affiliation(s)
- James D Otvos
- Laboratory Corporation of America(®) Holdings (LabCorp), Morrisville, NC, USA
| | - John R Guyton
- Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| | - Margery A Connelly
- Laboratory Corporation of America(®) Holdings (LabCorp), Morrisville, NC, USA
| | | | - Vera Bittner
- Department of Cardiology, Prevention and Imaging, University of Alabama, Birmingham, AL, USA
| | | | | | | | - Joseph B Muhlestein
- Intermountain Medical Center, Murray, UT, USA; University of Utah, Salt Lake City, UT, USA
| | - William E Boden
- VA New England Healthcare System, Bedford, MA, USA; Massachusetts Veterans Epidemiology, Research, and Informatics Center (MAVERIC), and Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
426
|
Shoji M, Takemoto M, Kobayashi K, Shoji T, Mori S, Sagara JI, Kurosawa H, Hirayama Y, Sakamoto K, Ishikawa T, Koshizaka M, Maezawa Y, Yokote K. Serum podocalyxin levels correlate with carotid intima media thickness, implicating its role as a novel biomarker for atherosclerosis. Sci Rep 2018; 8:245. [PMID: 29321582 PMCID: PMC5762903 DOI: 10.1038/s41598-017-18647-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 12/15/2017] [Indexed: 12/05/2022] Open
Abstract
Podocalyxin is a cell surface sialomucin, which is expressed in not only glomerular podocytes but also vascular endothelial cells. Urinary podocalyxin is used as a marker for glomerular disease. However, there are no reports describing serum podocalyxin (s-Podxl) levels. Therefore, the association between s-Podxl levels and clinical parameters were examined with 52 patients. s-Podxl level was evaluated using enzyme-linked immunosorbent assay. The median s-Podxl level was 14.2 ng/dL (interquartile range: 10.8–22.2 ng/dL). There were significant correlations (correlation coefficient: r > 0.2) of s-Podxl levels with carotid intima media thickness (IMT) (r = 0.30, p = 0.0307). Multiple logistic regression analysis showed that s-Podxl levels remained significantly associated with carotid IMT > 1 mm (OR: 1.15; 95% CI 1.02–1.31, p = 0.026) after adjustments for traditional cardiovascular risk factors such as age, sex, current smoking status, hypertension, dyslipidemias, and diabetes. In conclusion, s-Podxl is independently associated with carotid IMT and might be used as a novel biomarker for cardiovascular disease.
Collapse
Affiliation(s)
- Mayumi Shoji
- Department of Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan.,Department of Medicine, Division of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan
| | - Minoru Takemoto
- Department of Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan. .,School of Medicine, International University of Health and Welfare, Department of Diabetes, Metabolism and Endocrinology, 4-3 Kozunomori, Narita-shi, Chiba, 286-8686, Japan.
| | - Kazuki Kobayashi
- Department of Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan.,Asahi Chuo Hospital, 1326, Ino, Asahi-shi, Chiba, 289-2511, Japan
| | - Toshihiro Shoji
- Department of Cardiology, Chiba Emergency Medical Center, Chiba, 3-32-1, Isobe, Mihama-ku, Chiba-shi, Chiba, 261-0012, Japan
| | - Satoka Mori
- Life Inovation Research Institute, Denka Co., Ltd, 3-5-1, Asahi-Machi, Machida-City, Tokyo, 194-0023, Japan
| | - Jun-Ichi Sagara
- Life Inovation Research Institute, Denka Co., Ltd, 3-5-1, Asahi-Machi, Machida-City, Tokyo, 194-0023, Japan
| | - Hiroyuki Kurosawa
- R&D Center, Denka Seikne Co., Ltd, 1359-1, Kagamida, Kigoshi, Gosen-City, Niigata, 959-1695, Japan
| | - Yoshiaki Hirayama
- R&D Center, Denka Seikne Co., Ltd, 1359-1, Kagamida, Kigoshi, Gosen-City, Niigata, 959-1695, Japan
| | - Kenichi Sakamoto
- Department of Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan.,Department of Medicine, Division of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan
| | - Takahiro Ishikawa
- Department of Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan.,Department of Medicine, Division of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan
| | - Masaya Koshizaka
- Department of Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan.,Department of Medicine, Division of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan
| | - Yoshiro Maezawa
- Department of Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan.,Department of Medicine, Division of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan
| | - Koutaro Yokote
- Department of Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan.,Department of Medicine, Division of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan
| |
Collapse
|
427
|
Baumann R, Gube M, Markert A, Davatgarbenam S, Kossack V, Gerhards B, Kraus T, Brand P. Systemic serum amyloid A as a biomarker for exposure to zinc and/or copper-containing metal fumes. JOURNAL OF EXPOSURE SCIENCE & ENVIRONMENTAL EPIDEMIOLOGY 2018; 28:84-91. [PMID: 28176762 DOI: 10.1038/jes.2016.86] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 12/06/2016] [Indexed: 06/06/2023]
Abstract
Zinc- and copper-containing welding fumes increase systemic C-reactive protein (CRP). The aim of this study was to investigate the performance of the biomarkers serum amyloid A (SAA) and soluble vascular cell adhesion molecule-1 (VCAM-1) in this regard. Fifteen male subjects were exposed under controlled conditions to welding fumes containing either zinc, or copper, or copper and zinc for 6 h. Plasma samples were collected before, 6 and 24 h after start of exposure and biomarkers therein were measured by electrochemiluminescent assay. For each exposure, systemic concentrations of systemic SAA, but not VCAM-1, increased significantly at 24 h after exposure start compared with baseline ("copper only": P=0.0005, "zinc only": P=0.027, "copper and zinc": P=0.001). SAA showed a wider range of concentrations than did CRP and its levels increased up to 19-fold after welding fume exposure. The recognition of copper as a potential harmful component in welding fumes, also independent from zinc, deserves further consideration. SAA might represent a new sensitive biomarker for potential subclinical sterile inflammation after inhalation of copper- and/or zinc-containing welding fumes. As elevations of CRP and SAA protein have both been linked to a higher risk for cardiovascular disease, these findings might particularly be important for long-term welders.
Collapse
Affiliation(s)
- R Baumann
- Institute for Occupational and Social Medicine, Aachen University of Technology, Pauwelsstr, Germany
| | - M Gube
- Institute for Occupational and Social Medicine, Aachen University of Technology, Pauwelsstr, Germany
| | - A Markert
- Institute for Occupational and Social Medicine, Aachen University of Technology, Pauwelsstr, Germany
| | - S Davatgarbenam
- Institute for Occupational and Social Medicine, Aachen University of Technology, Pauwelsstr, Germany
| | - V Kossack
- Institute for Occupational and Social Medicine, Aachen University of Technology, Pauwelsstr, Germany
| | - B Gerhards
- ISF - Welding and Joining Institute, Aachen University of Technology, Pontstraße, Germany
| | - T Kraus
- Institute for Occupational and Social Medicine, Aachen University of Technology, Pauwelsstr, Germany
| | - P Brand
- Institute for Occupational and Social Medicine, Aachen University of Technology, Pauwelsstr, Germany
| |
Collapse
|
428
|
Hou J, Sun H, Guo Y, Zhou Y, Yin W, Xu T, Cheng J, Chen W, Yuan J. Associations between urinary monohydroxy polycyclic aromatic hydrocarbons metabolites and Framingham Risk Score in Chinese adults with low lung function. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 147:1002-1009. [PMID: 29976002 DOI: 10.1016/j.ecoenv.2017.09.058] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/18/2017] [Accepted: 09/20/2017] [Indexed: 06/08/2023]
Abstract
Previous studies have reported an association of exposure to polycyclic aromatic hydrocarbons (PAHs) with lung function decline or cardiovascular diseases, or reduced lung function with 10-year cardiovascular (CV) risk. We analyzed risk factors for the 10-year Framingham CV risk using multiple logistic regression, and examined the mediational effect of reduced lung function on the association between exposure to PAHs and FRS using the post-exploratory structural equation modeling. Participants (n = 2268) were drawn from the Wuhan residents at baseline from the Wuhan-Zhuhai Cohort Study. They completed the physical examination, measurements of lung function and urinary monohydroxylated-PAHs (OH-PAHs). In all individuals, we found a dose-response relationship of PAHs exposure, forced expiratory volume in 1s (FEV1) or forced vital capacity (FVC) with the 10-year CV risk. The proportions of FEV1 and FVC mediation effects in association of PAH exposure with the10-year CV risk were 35% and 24%, respectively. The findings indicated that PAHs exposure or reduced lung function increased the 10-year CV risk. Impaired lung function may partly contribute to increase in the 10-year CV risk regarding exposure to PAHs.
Collapse
Affiliation(s)
- Jian Hou
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China
| | - Huizhen Sun
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China
| | - Yanjun Guo
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China
| | - Yun Zhou
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China
| | - Wenjun Yin
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China
| | - Tian Xu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China
| | - Juan Cheng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China
| | - Weihong Chen
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China.
| | - Jing Yuan
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China.
| |
Collapse
|
429
|
|
430
|
Gan W, Yi Y, Fu Y, Huang J, Lu Z, Jing C, Fan J, Zhou J, Qiu S. Fibrinogen and C-reactive protein score is a prognostic index for patients with hepatocellular carcinoma undergoing curative resection: a prognostic nomogram study. J Cancer 2018; 9:148-156. [PMID: 29290780 PMCID: PMC5743722 DOI: 10.7150/jca.22246] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 10/19/2017] [Indexed: 01/03/2023] Open
Abstract
Background: While curative resection is the established strategy for Hepatocellular carcinoma (HCC) patients, the prognosis still remains poor, and the efficiency of existing prediction models is unsatisfactory. Therefore, we aimed to develop a credible and easy-to-use prognostic index for patients with HCC undergoing curative therapy. Methods: A total of 768 patients with HCC, who underwent curative resection from December 2010 to June 2012 in Zhongshan Hospital, were divided into a training cohort with 616 patients and a validating cohort of 152 patients at a ratio of 4 to 1 by random allocation. Then, a retrospective cohort study was conducted to identify effective prognostic indexes. Results: FC-score, which incorporates fibrinogen and C-reactive protein, was established. In the multivariate analysis for OS and RFS, FC-score has shown to be a significant independent prognostic index in both the training cohort and validation cohort. Furthermore, the C-index of the FC-score for OS and RFS were 0.698 and 0.594 respectively, which were superior to other inflammation systems such as IBI, IBS, and GPS. Then, we developed a novel nomogram, which integrated FC-score into the conventional BCLC staging system. This new nomogram gave rise to a new C-index of 0.746 (95%CI: 0.743-0.749) for OS, and 0.654 (95%CI: 0.652-0.656) for RFS. The calibration curve and decision curve analysis indicated that our nomogram was highly consistent between predicted and actual observations. Conclusions: FC-score represents a novel, convenient, reliable, and accurate prognostic predictor for both OS and RFS in HCC patients undergoing curative therapy.
Collapse
Affiliation(s)
- Wei Gan
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, Fudan University, Key Laboratory for Carcinogenesis & Cancer Invasion, The Chinese Ministry of Education, Shanghai, China
| | - Yong Yi
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, Fudan University, Key Laboratory for Carcinogenesis & Cancer Invasion, The Chinese Ministry of Education, Shanghai, China
| | - Yipeng Fu
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, Fudan University, Key Laboratory for Carcinogenesis & Cancer Invasion, The Chinese Ministry of Education, Shanghai, China
| | - Jinlong Huang
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, Fudan University, Key Laboratory for Carcinogenesis & Cancer Invasion, The Chinese Ministry of Education, Shanghai, China
| | - Zhufeng Lu
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, Fudan University, Key Laboratory for Carcinogenesis & Cancer Invasion, The Chinese Ministry of Education, Shanghai, China.,Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chuyu Jing
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, Fudan University, Key Laboratory for Carcinogenesis & Cancer Invasion, The Chinese Ministry of Education, Shanghai, China
| | - Jia Fan
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, Fudan University, Key Laboratory for Carcinogenesis & Cancer Invasion, The Chinese Ministry of Education, Shanghai, China
| | - Jian Zhou
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, Fudan University, Key Laboratory for Carcinogenesis & Cancer Invasion, The Chinese Ministry of Education, Shanghai, China
| | - Shuangjian Qiu
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, Fudan University, Key Laboratory for Carcinogenesis & Cancer Invasion, The Chinese Ministry of Education, Shanghai, China.,Biomedical Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
431
|
Eelen G, de Zeeuw P, Treps L, Harjes U, Wong BW, Carmeliet P. Endothelial Cell Metabolism. Physiol Rev 2018; 98:3-58. [PMID: 29167330 PMCID: PMC5866357 DOI: 10.1152/physrev.00001.2017] [Citation(s) in RCA: 384] [Impact Index Per Article: 54.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 06/19/2017] [Accepted: 06/22/2017] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells (ECs) are more than inert blood vessel lining material. Instead, they are active players in the formation of new blood vessels (angiogenesis) both in health and (life-threatening) diseases. Recently, a new concept arose by which EC metabolism drives angiogenesis in parallel to well-established angiogenic growth factors (e.g., vascular endothelial growth factor). 6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3-driven glycolysis generates energy to sustain competitive behavior of the ECs at the tip of a growing vessel sprout, whereas carnitine palmitoyltransferase 1a-controlled fatty acid oxidation regulates nucleotide synthesis and proliferation of ECs in the stalk of the sprout. To maintain vascular homeostasis, ECs rely on an intricate metabolic wiring characterized by intracellular compartmentalization, use metabolites for epigenetic regulation of EC subtype differentiation, crosstalk through metabolite release with other cell types, and exhibit EC subtype-specific metabolic traits. Importantly, maladaptation of EC metabolism contributes to vascular disorders, through EC dysfunction or excess angiogenesis, and presents new opportunities for anti-angiogenic strategies. Here we provide a comprehensive overview of established as well as newly uncovered aspects of EC metabolism.
Collapse
Affiliation(s)
- Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Pauline de Zeeuw
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Ulrike Harjes
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Brian W Wong
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| |
Collapse
|
432
|
Steptoe A, Kivimäki M, Lowe G, Rumley A, Hamer M. Blood Pressure and Fibrinogen Responses to Mental Stress as Predictors of Incident Hypertension over an 8-Year Period. Ann Behav Med 2017; 50:898-906. [PMID: 27401000 PMCID: PMC5126198 DOI: 10.1007/s12160-016-9817-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background Heightened blood pressure (BP) responses to mental stress predict raised BP levels over subsequent years, but evidence for associations with incident hypertension is limited, and the significance of inflammatory responses is uncertain. Purpose We investigated the relationship between BP and plasma fibrinogen responses to stress and incident hypertension over an average 8-year follow-up. Method Participants were 636 men and women (mean age 59.1 years) from the Whitehall II epidemiological cohort with no history of cardiovascular disease and hypertension. They performed standardized behavioral tasks (color/word conflict and mirror tracing), and hypertension was defined by clinic measures and medication status. Results Of participants in the highest systolic BP reactivity tertile, 29.3 % became hypertensive over the follow-up period compared with 16.5 % of those in the lowest tertile, with an odds ratio of 2.02 (95 % CI 1.17–3.88, p = 0.012) after adjustment for age, sex, grade of employment, body mass index, smoking, alcohol consumption, physical activity, follow-up time, subjective stress response, perceived task difficulty, perceived task engagement, and baseline BP. Similar associations were observed for diastolic BP reactivity (odds ratio 2.05, 95 % CI 1.23–3.40, p = 0.006) and for impaired systolic BP post-stress recovery (odds ratio 2.06, 95 % CI 1.19–3.57, p = 0.010). Fibrinogen reactions to tasks also predicted future hypertension in women (odds ratio 2.64, 95 % CI 1.11–6.30, p = 0.029) but not men. Conclusions These data suggest that heightened cardiovascular and inflammatory reactivity to mental stress is associated with hypertension risk, and may be a mechanism through which psychosocial factors impact on the development of hypertension.
Collapse
Affiliation(s)
- Andrew Steptoe
- Department of Epidemiology and Public Health, University College London, 1-19 Torrington Place, London, WC1E 6BT, UK.
| | - Mika Kivimäki
- Department of Epidemiology and Public Health, University College London, 1-19 Torrington Place, London, WC1E 6BT, UK
| | - Gordon Lowe
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland
| | - Ann Rumley
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland
| | - Mark Hamer
- National Centre for Sport & Exercise Medicine, Loughborough University, Loughborough, UK
| |
Collapse
|
433
|
Khosrow-Khavar F, Filion KB, Al-Qurashi S, Torabi N, Bouganim N, Suissa S, Azoulay L. Cardiotoxicity of aromatase inhibitors and tamoxifen in postmenopausal women with breast cancer: a systematic review and meta-analysis of randomized controlled trials. Ann Oncol 2017; 28:487-496. [PMID: 27998966 DOI: 10.1093/annonc/mdw673] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Aromatase inhibitors (AIs) have been associated with cardiovascular disease in adjuvant randomized controlled trials (RCTs) comparing these drugs to tamoxifen. However, it is unclear whether this risk is real or due to cardioprotective effects of tamoxifen. To address this question, we conducted a systematic review and meta-analysis of all RCTs of AIs and tamoxifen in adjuvant and extended adjuvant setting. Patients and methods We searched PubMed, Embase (OVID), Cochrane CENTRAL, WHO International Clinical Trials Registry Platform, and ClinicalTrials.gov from inception to June 2016 for all RCTs comparing cardiovascular and cerebrovascular safety of AIs to tamoxifen, AIs to placebo or no-treatment, or tamoxifen to placebo or no-treatment in the adjuvant or extended adjuvant setting. Relative risks (RRs) were pooled using DerSimonian and Laird random-effects models with analyses stratified by RCT design. Results A total of 19 RCTs were included in the meta-analysis (n = 62 345). In the adjuvant setting, AIs were associated with a 19% (RR: 1.19, 95% confidence interval [CI]: 1.07-1.34) increased risk of cardiovascular events compared with tamoxifen. AIs were not associated with an increased risk compared with placebo in the extended-adjuvant setting (RR: 1.01, 95% CI: 0.85-1.20). In the adjuvant setting, tamoxifen was associated with a 33% (RR: 0.67, 95% CI: 0.45-0.98) decreased risk compared with placebo or no-treatment. The results from extended adjuvant RCTs comparing tamoxifen to placebo were inconclusive but suggestive of a small protective effect (RR: 0.91, 95% CI: 0.77-1.07). Conclusions The increased risk of cardiovascular events with AIs relative to tamoxifen is likely the result of cardioprotective effects of the latter. This new evidence should be considered when assessing the benefits and risks of AIs in the treatment of breast cancer.
Collapse
Affiliation(s)
- F Khosrow-Khavar
- Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Canada
| | - K B Filion
- Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Canada
| | - S Al-Qurashi
- Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - N Torabi
- McGill Library, McGill University, Montreal, Quebec, Canada
| | - N Bouganim
- Department of Oncology, Cedar Cancer Center, McGill University Health Center, Montreal, Canada
| | - S Suissa
- Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Canada
| | - L Azoulay
- Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Canada
| |
Collapse
|
434
|
Paraboschi EM, Duga S, Asselta R. Fibrinogen as a Pleiotropic Protein Causing Human Diseases: The Mutational Burden of Aα, Bβ, and γ Chains. Int J Mol Sci 2017; 18:E2711. [PMID: 29240685 PMCID: PMC5751312 DOI: 10.3390/ijms18122711] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/11/2017] [Accepted: 12/13/2017] [Indexed: 12/19/2022] Open
Abstract
Fibrinogen is a highly pleiotropic protein that is involved in the final step of the coagulation cascade, wound healing, inflammation, and angiogenesis. Heterozygous mutations in Aα, Bβ, or γ fibrinogen-chain genes (FGA, FGB, FGG) have been described as being responsible for fibrinogen deficiencies (hypofibrinogenemia, hypo-dysfibrinogenemia, dysfibrinogenemia) and for more rare conditions, such as fibrinogen storage disease and hereditary renal amyloidosis. Instead, biallelic mutations have been associated with afibrinogenemia/severe hypofibrinogenemia, i.e., the severest forms of fibrinogen deficiency, affecting approximately 1-2 cases per million people. However, the "true" prevalence for these conditions on a global scale is currently not available. Here, we defined the mutational burden of the FGA, FGB, and FGG genes, and estimated the prevalence of inherited fibrinogen disorders through a systematic analysis of exome/genome data from ~140,000 individuals belonging to the genome Aggregation Database. Our analysis showed that the world-wide prevalence for recessively-inherited fibrinogen deficiencies could be 10-fold higher than that reported so far (prevalence rates vary from 1 in 10⁶ in East Asians to 24.5 in 10⁶ in non-Finnish Europeans). The global prevalence for autosomal-dominant fibrinogen disorders was estimated to be ~11 in 1000 individuals, with heterozygous carriers present at a frequency varying from 3 every 1000 individuals in Finns, to 1-2 every 100 individuals among non-Finnish Europeans and Africans/African Americans. Our analysis also allowed for the identification of recurrent (i.e., FGG-p.Ala108Gly, FGG-Thr47Ile) or ethnic-specific mutations (e.g., FGB-p.Gly103Arg in Admixed Americans, FGG-p.Ser245Phe in Africans/African Americans).
Collapse
Affiliation(s)
- Elvezia Maria Paraboschi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy.
| | - Stefano Duga
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy.
- Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano, Milan, Italy.
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy.
- Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano, Milan, Italy.
| |
Collapse
|
435
|
Cerit L. Is There Any Link Among C-Reactive Protein, Right Ventricular Function, and Postoperative Atrial Fibrillation? J Cardiothorac Vasc Anesth 2017; 32:e52-e53. [PMID: 29229254 DOI: 10.1053/j.jvca.2017.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Levent Cerit
- Department of Cardiology, Near East University, Nicosia, Cyprus
| |
Collapse
|
436
|
Ting PC, Chou AH, Wu VCC, Chen SW. Reply to "Is There Any Link Among C-Reactive Protein, Right Ventricular Function, and Postoperative Atrial Fibrillation?". J Cardiothorac Vasc Anesth 2017; 32:e53-e54. [PMID: 29229251 DOI: 10.1053/j.jvca.2017.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Pei-Chi Ting
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - An-Hsun Chou
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | | | - Shao-Wei Chen
- Division of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
437
|
Sweeting MJ, Barrett JK, Thompson SG, Wood AM. The use of repeated blood pressure measures for cardiovascular risk prediction: a comparison of statistical models in the ARIC study. Stat Med 2017; 36:4514-4528. [PMID: 27730661 PMCID: PMC5724484 DOI: 10.1002/sim.7144] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 09/01/2016] [Accepted: 09/18/2016] [Indexed: 01/22/2023]
Abstract
Many prediction models have been developed for the risk assessment and the prevention of cardiovascular disease in primary care. Recent efforts have focused on improving the accuracy of these prediction models by adding novel biomarkers to a common set of baseline risk predictors. Few have considered incorporating repeated measures of the common risk predictors. Through application to the Atherosclerosis Risk in Communities study and simulations, we compare models that use simple summary measures of the repeat information on systolic blood pressure, such as (i) baseline only; (ii) last observation carried forward; and (iii) cumulative mean, against more complex methods that model the repeat information using (iv) ordinary regression calibration; (v) risk-set regression calibration; and (vi) joint longitudinal and survival models. In comparison with the baseline-only model, we observed modest improvements in discrimination and calibration using the cumulative mean of systolic blood pressure, but little further improvement from any of the complex methods. © 2016 The Authors. Statistics in Medicine Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- Michael J Sweeting
- Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge, U.K
| | - Jessica K Barrett
- Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge, U.K
| | - Simon G Thompson
- Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge, U.K
| | - Angela M Wood
- Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge, U.K
| |
Collapse
|
438
|
Casula M, Montecucco F, Bonaventura A, Liberale L, Vecchié A, Dallegri F, Carbone F. Update on the role of Pentraxin 3 in atherosclerosis and cardiovascular diseases. Vascul Pharmacol 2017; 99:1-12. [PMID: 29051088 DOI: 10.1016/j.vph.2017.10.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 10/11/2017] [Accepted: 10/15/2017] [Indexed: 12/12/2022]
Abstract
Pentraxin 3 (PTX3) is an acute-phase protein that was recently demonstrated to play pleiotropic activities in cardiovascular (CV) diseases. Tumor necrosis factor and interleukins up-regulates PTX3 transcription in different cell types (i.e. endothelial cells, phagocytes, smooth muscle cells, fibroblasts and glial cells) involved in atherogenesis. By interacting with numerous ligands, PTX3 acts as a modulatory molecule of complement system, inflammatory response, angiogenesis, and vascular/tissue remodeling. Experimental data point to a beneficial role of PTX3 in atherosclerotic plaque development and vulnerability. Animal studies indicated a protective role of PTX3 signaling in ischemic/reperfusion injury and failing heart. Clinical studies have so far provided contrasting results, highlighting a debated role of PTX3 as an active mediator of endothelial dysfunction, atherosclerotic plaque vulnerability and worse outcome after ischemic events. Therefore, substantial evidence suggests a dual role of PTX3 as modulator or amplifiers of the innate immune response. The final result of PTX3 activation might be determined by a fine tuning of time, space and environmental signals. The aim of this review is to provide an overview of biological properties of PTX3 in CV diseases and to discuss the ability of PTX3 to act as a crossroad between pro- and anti-inflammatory pathways.
Collapse
Affiliation(s)
- Matteo Casula
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; Ospedale Policlinico San Martino, 10 Largo Benzi, 16132 Genoa, Italy; Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 9 viale Benedetto XV, 16132 Genoa, Italy
| | - Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, CH-8952 Schlieren, Switzerland
| | - Alessandra Vecchié
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; Ospedale Policlinico San Martino, 10 Largo Benzi, 16132 Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.
| |
Collapse
|
439
|
Lussana F, Rambaldi A. Inflammation and myeloproliferative neoplasms. J Autoimmun 2017; 85:58-63. [DOI: 10.1016/j.jaut.2017.06.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 01/14/2023]
|
440
|
Lahoud R, Chongthammakun V, Wu Y, Hawwa N, Brennan DM, Cho L. Comparing SF-36® scores versus biomarkers to predict mortality in primary cardiac prevention patients. Eur J Intern Med 2017. [PMID: 28625611 DOI: 10.1016/j.ejim.2017.05.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Risk stratification plays an important role in evaluating patients with no known cardiovascular disease (CVD). Few studies have investigated health-related quality of life questionnaires such as the Medical Outcomes Study Short Form-36 (SF-36®) as predictive tools for mortality, particularly in direct comparison with biomarkers. Our objective is to measure the relative effectiveness of SF-36® scores in predicting mortality when compared to traditional and novel biomarkers in a primary prevention population. METHODS 7056 patients evaluated for primary cardiac prevention between January 1996 and April 2011 were included in this study. Patient characteristics included medical history, SF-36® questionnaire and a laboratory panel (total cholesterol, triglycerides, HDL, LDL, ApoA, ApoB, ApoA1/ApoB ratio, homocysteine, lipoprotein (a), fibrinogen, hsCRP, uric acid and urine ACR). The primary outcome was all-cause mortality. RESULTS A low SF-36® physical score independently predicted a 6-fold increase in death at 8years (above vs. below median Hazard Ratio [95% confidence interval] 5.99 [3.86-9.35], p<0.001). In a univariate analysis, SF-36® physical score had a c-index of 0.75, which was superior to that of all the biomarkers. It also carried incremental predictive ability when added to non-laboratory risk factors (Net Reclassification Index=59.9%), as well as Framingham risk score components (Net Reclassification Index=61.1%). Biomarkers added no incremental predictive value to a non-laboratory risk factor model when combined to SF-36 physical score. CONCLUSION The SF-36® physical score is a reliable predictor of mortality in patients without CVD, and outperformed most studied traditional and novel biomarkers. In an era of rising healthcare costs, the SF-36® questionnaire could be used as an adjunct simple and cost-effective predictor of mortality to current predictors.
Collapse
Affiliation(s)
- Rony Lahoud
- Cleveland Clinic Foundation, Heart Vascular Institute, Department of Cardiology, Cleveland, OH, United States
| | | | - Yuping Wu
- Cleveland Clinic Foundation, Heart Vascular Institute, Department of Cardiology, Cleveland, OH, United States
| | - Nael Hawwa
- Cleveland Clinic Foundation, Heart Vascular Institute, Department of Cardiology, Cleveland, OH, United States
| | - Danielle M Brennan
- C5Research, Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, United States
| | - Leslie Cho
- Cleveland Clinic Foundation, Heart Vascular Institute, Department of Cardiology, Cleveland, OH, United States.
| |
Collapse
|
441
|
Saleh AD, Kwok B, Brown JS, Hurst JR. Correlates and assessment of excess cardiovascular risk in bronchiectasis. Eur Respir J 2017; 50:50/5/1701127. [PMID: 29167299 DOI: 10.1183/13993003.01127-2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/16/2017] [Indexed: 12/20/2022]
Abstract
Patients with bronchiectasis are at increased risk of cardiovascular disease. We aimed to identify factors associated with elevated cardiovascular risk in bronchiectasis, measured using aortic stiffness and cardiac biomarkers. In addition, we sought to compare these direct measures against calculated QRISK2 scores.Aortic stiffness, cardiac biomarkers and systemic inflammation were measured in 101 adults with stable bronchiectasis. In addition, clinical and demographic data were collected to allow calculation of QRISK2 score and the bronchiectasis severity index (BSI) for each patient.The BSI score correlated with measured cardiovascular risk assessments, partly due to greater exacerbation frequency and lower forced expiratory volume in 1 s. Pulse-wave velocity was significantly higher in frequent exacerbators (≥3 events·year-1) than infrequent exacerbators (<3 events·year-1; 10.5 versus 9.2 m·s-1, p=0.01). In addition, frequent exacerbators had elevated serum C-reactive protein concentration, suggesting increased systemic inflammation (4.8 versus 2.2 mg·L-1, p=0.005). QRISK2 systematically underestimated cardiovascular risk in this population (median change in relative risk 1.29). Underestimation was associated with frequent exacerbations and male sex.Patients with bronchiectasis have greater cardiovascular risk than published reference populations. Excess cardiovascular risk is associated with exacerbation frequency and impaired lung function. Cardiovascular risk assessment in bronchiectasis should be individualised, as calculation tools are likely to underestimate the risk in this population.
Collapse
Affiliation(s)
- Aarash D Saleh
- Centre for Inflammation and Tissue Repair, UCL Respiratory Medicine, Royal Free Campus, University College London, London, UK
| | - Bessie Kwok
- Centre for Inflammation and Tissue Repair, UCL Respiratory Medicine, Royal Free Campus, University College London, London, UK
| | - Jeremy S Brown
- Centre for Inflammation and Tissue Repair, UCL Respiratory Medicine, Royal Free Campus, University College London, London, UK
| | - John R Hurst
- Centre for Inflammation and Tissue Repair, UCL Respiratory Medicine, Royal Free Campus, University College London, London, UK
| |
Collapse
|
442
|
Rumley A, Lowe G. The relevance of coagulation in cardiovascular disease: what do the biomarkers tell us? Thromb Haemost 2017; 112:860-7. [DOI: 10.1160/th14-03-0199] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 07/22/2014] [Indexed: 11/05/2022]
Abstract
SummarySeveral haemostatic factors have been associated with incident arterial cardiovascular disease in prospective studies and meta-analyses. Plasma fibrinogen shows a strong and consistent association with risk; however, this may reflect its inflammatory marker status, and causality remains to be proven. The common haemostatic gene polymorphisms for factor II, factor V and the von Willebrand factor: Factor VIII (non-O blood group) show significant associations with coronary heart disease (CHD) risk, consistent with potential causality. Increased D-dimer and t-PA antigen levels are associated with CHD risk, suggesting roles for coagulation activation and endothelial disturbance. There is little evidence for associations with CVD with other haemostatic factors.
Collapse
|
443
|
Altes P, Perez P, Esteban C, Sánchez Muñoz-Torrero JF, Aguilar E, García-Díaz AM, Álvarez LR, Jiménez PE, Sahuquillo JC, Monreal M. Raised Fibrinogen Levels and Outcome in Outpatients With Peripheral Artery Disease. Angiology 2017; 69:507-512. [PMID: 29113452 DOI: 10.1177/0003319717739720] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The influence of raised fibrinogen levels on outcome in stable outpatients with peripheral arterial disease (PAD) has not been consistently investigated. We used data from the Factores de Riesgo y ENfermedad Arterial (FRENA) registry to compare ischemic events, major bleeding, and mortality in stable outpatients with PAD, according to their baseline plasma fibrinogen levels. Of 1363 outpatients with PAD recruited in FRENA, 558 (41%) had fibrinogen levels >450 mg/100 mL. Over 18 months, 43 patients presented with acute myocardial infarction, 37 had an ischemic stroke, 51 underwent limb amputation, 19 had major bleeding, and 90 died. Compared to patients with normal levels, those with raised fibrinogen levels had an over 2-fold higher rate of ischemic stroke (rate ratio [RR]: 2.30; 95% confidence interval [CI]: 1.19-4.59), limb amputation (RR: 2.58; 95% CI: 1.46-4.67), or death (RR: 2.27; 95% CI: 1.49-3.51) and an over 3-fold higher rate of major bleeding (RR: 3.90; 95% CI: 1.45-12.1). On multivariate analysis, patients with raised fibrinogen levels had an increased risk of developing subsequent ischemic events (hazard ratio [HR]: 1.61; 95% CI: 1.11-2.32) and major bleeding (HR: 3.42; 95% CI: 1.22-9.61). Stable outpatients with PAD and raised plasma fibrinogen levels had increased rates of subsequent ischemic events and major bleeding.
Collapse
Affiliation(s)
- Pere Altes
- 1 Department of Vascular Surgery, Hospital Universitari Germans Trias i Pujol, Badalona, Facultat de Medicina, Universitat Autònoma de Barcelona, Spain
| | - Paulina Perez
- 1 Department of Vascular Surgery, Hospital Universitari Germans Trias i Pujol, Badalona, Facultat de Medicina, Universitat Autònoma de Barcelona, Spain
| | - Carlos Esteban
- 1 Department of Vascular Surgery, Hospital Universitari Germans Trias i Pujol, Badalona, Facultat de Medicina, Universitat Autònoma de Barcelona, Spain
| | | | - Eduardo Aguilar
- 3 Department of Internal Medicine, Hospital de Alcañiz, Alcañiz, Teruel, Spain
| | | | - Lorenzo Ramón Álvarez
- 5 Department of Vascular Surgery, CST-Hospital de Terrassa, Terrassa, Barcelona, Spain
| | | | | | - Manuel Monreal
- 8 Department of Internal Medicine, Hospital Universitari Germans Trias i Pujol, Badalona, Facultat de Medicina, Universitat Autònoma de Barcelona, Spain
| | | |
Collapse
|
444
|
Ghasemzedah N, Hayek SS, Ko YA, Eapen DJ, Patel RS, Manocha P, Al Kassem H, Khayata M, Veledar E, Kremastinos D, Thorball CW, Pielak T, Sikora S, Zafari AM, Lerakis S, Sperling L, Vaccarino V, Epstein SE, Quyyumi AA. Pathway-Specific Aggregate Biomarker Risk Score Is Associated With Burden of Coronary Artery Disease and Predicts Near-Term Risk of Myocardial Infarction and Death. Circ Cardiovasc Qual Outcomes 2017; 10:CIRCOUTCOMES.115.001493. [PMID: 28280039 DOI: 10.1161/circoutcomes.115.001493] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 01/11/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Inflammation, coagulation, and cell stress contribute to atherosclerosis and its adverse events. A biomarker risk score (BRS) based on the circulating levels of biomarkers C-reactive protein, fibrin degradation products, and heat shock protein-70 representing these 3 pathways was a strong predictor of future outcomes. We investigated whether soluble urokinase plasminogen activator receptor (suPAR), a marker of immune activation, is predictive of outcomes independent of the aforementioned markers and whether its addition to a 3-BRS improves risk reclassification. METHODS AND RESULTS C-reactive protein, fibrin degradation product, heat shock protein-70, and suPAR were measured in 3278 patients undergoing coronary angiography. The BRS was calculated by counting the number of biomarkers above a cutoff determined using the Youden's index. Survival analyses were performed using models adjusted for traditional risk factors. A high suPAR level ≥3.5 ng/mL was associated with all-cause death and myocardial infarction (hazard ratio, 1.83; 95% confidence interval, 1.43-2.35) after adjustment for risk factors, C-reactive protein, fibrin degradation product, and heat shock protein-70. Addition of suPAR to the 3-BRS significantly improved the C statistic, integrated discrimination improvement, and net reclassification index for the primary outcome. A BRS of 1, 2, 3, or 4 was associated with a 1.81-, 2.59-, 6.17-, and 8.80-fold increase, respectively, in the risk of death and myocardial infarction. The 4-BRS was also associated with severity of coronary artery disease and composite end points. CONCLUSIONS SuPAR is independently predictive of adverse outcomes, and its addition to a 3-BRS comprising C-reactive protein, fibrin degradation product, and heat shock protein-70 improved risk reclassification. The clinical utility of using a 4-BRS for risk prediction and management of patients with coronary artery disease warrants further study.
Collapse
Affiliation(s)
- Nima Ghasemzedah
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - Salim S Hayek
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - Yi-An Ko
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - Danny J Eapen
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - Riyaz S Patel
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - Pankaj Manocha
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - Hatem Al Kassem
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - Mohamed Khayata
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - Emir Veledar
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - Dimitrios Kremastinos
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - Christian W Thorball
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - Tomasz Pielak
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - Sergey Sikora
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - A Maziar Zafari
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - Stamatios Lerakis
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - Laurence Sperling
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - Viola Vaccarino
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - Stephen E Epstein
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.)
| | - Arshed A Quyyumi
- From the Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA (N.G., S.S.H., D.J.E., R.S.P., P.M., H.A.K., M.K., E.V., A.M.Z., S.L., L.S., V.V., A.A.Q.); Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA (Y.-A.K.); Institute of Cardiovascular Science, University College London, United Kingdom (R.S.P.); Division of Cardiology, Atlanta VA Medical Center, GA (A.M.Z.); Department of Biostatistics, Florida International University, Miami (E.V.); Department of Cardiology, University of Athens School of Medicine, Greece (D.K.); Clinical Research Centre, Copenhagen University Hospital, Denmark (C.W.T., T.P.); Stemedica Cell Technologies, Inc., San Diego, CA (S.S.); Department of Epidemiology, Emory University, Atlanta, GA (V.V.); and MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC (S.E.E.).
| |
Collapse
|
445
|
|
446
|
Carnethon MR, Pu J, Howard G, Albert MA, Anderson CAM, Bertoni AG, Mujahid MS, Palaniappan L, Taylor HA, Willis M, Yancy CW. Cardiovascular Health in African Americans: A Scientific Statement From the American Heart Association. Circulation 2017; 136:e393-e423. [PMID: 29061565 DOI: 10.1161/cir.0000000000000534] [Citation(s) in RCA: 784] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND PURPOSE Population-wide reductions in cardiovascular disease incidence and mortality have not been shared equally by African Americans. The burden of cardiovascular disease in the African American community remains high and is a primary cause of disparities in life expectancy between African Americans and whites. The objectives of the present scientific statement are to describe cardiovascular health in African Americans and to highlight unique considerations for disease prevention and management. METHOD The primary sources of information were identified with PubMed/Medline and online sources from the Centers for Disease Control and Prevention. RESULTS The higher prevalence of traditional cardiovascular risk factors (eg, hypertension, diabetes mellitus, obesity, and atherosclerotic cardiovascular risk) underlies the relatively earlier age of onset of cardiovascular diseases among African Americans. Hypertension in particular is highly prevalent among African Americans and contributes directly to the notable disparities in stroke, heart failure, and peripheral artery disease among African Americans. Despite the availability of effective pharmacotherapies and indications for some tailored pharmacotherapies for African Americans (eg, heart failure medications), disease management is less effective among African Americans, yielding higher mortality. Explanations for these persistent disparities in cardiovascular disease are multifactorial and span from the individual level to the social environment. CONCLUSIONS The strategies needed to promote equity in the cardiovascular health of African Americans require input from a broad set of stakeholders, including clinicians and researchers from across multiple disciplines.
Collapse
|
447
|
Sutin AR, Rust G, Robinson E, Daly M, Terracciano A. Parental perception of child weight and inflammation: Perceived overweight is associated with higher child c-reactive protein. Biol Psychol 2017; 130:50-53. [PMID: 29054818 DOI: 10.1016/j.biopsycho.2017.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 09/21/2017] [Accepted: 10/16/2017] [Indexed: 11/17/2022]
Abstract
Self-perceived overweight and weight discrimination are associated with inflammation in adulthood. We test whether there is an intergenerational association of parent perception of child overweight on higher levels of child c-reactive protein (CRP), a marker of inflammation implicated in stress. Data were from the National Health and Nutrition Examination Survey 2005-2014 (N=4988). Parents reported their perception of their child's weight; CRP was assayed from children's blood samples. Children whose parents perceived them as overweight had higher CRP levels than children who were perceived about the right weight; perceived underweight was also associated with higher CRP (F(2,4977)=9.23, p<.001). These associations were independent of the child's objective weight status and waist circumference and held when the sample was limited to children with objective overweight and obesity. These results suggest an intergenerational transfer of the psychological perception of body weight from parents to the inflammatory health of their child.
Collapse
Affiliation(s)
| | - George Rust
- Florida State University College of Medicine, United States
| | - Eric Robinson
- Institute of Psychology, Health & Society, University of Liverpool, United States
| | - Michael Daly
- Behavioural Science Centre, University of Stirling, United States; UCD Geary Institute, University College Dublin, United States
| | | |
Collapse
|
448
|
Nelson BW, Byrne ML, Simmons JG, Whittle S, Schwartz OS, O'Brien‐Simpson NM, Walsh KA, Reynolds EC, Allen NB. Adolescent temperament dimensions as stable prospective risk and protective factors for salivary C‐reactive protein. Br J Health Psychol 2017; 23:186-207. [DOI: 10.1111/bjhp.12281] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 09/21/2017] [Indexed: 12/23/2022]
Affiliation(s)
| | | | - Julian G. Simmons
- Melbourne School of Psychological Sciences The University of Melbourne Victoria Australia
- Melbourne Neuropsychiatry Centre Department of Psychiatry The University of Melbourne and Melbourne Health Victoria Australia
| | - Sarah Whittle
- Melbourne School of Psychological Sciences The University of Melbourne Victoria Australia
- Melbourne Neuropsychiatry Centre Department of Psychiatry The University of Melbourne and Melbourne Health Victoria Australia
| | - Orli S. Schwartz
- Melbourne Neuropsychiatry Centre Department of Psychiatry The University of Melbourne and Melbourne Health Victoria Australia
| | | | - Katrina A. Walsh
- Melbourne Dental School, Oral Health CRC The University of Melbourne Victoria Australia
| | - Eric C. Reynolds
- Melbourne Dental School, Oral Health CRC The University of Melbourne Victoria Australia
| | - Nicholas B. Allen
- Department of Psychology University of Oregon Eugene Oregon USA
- Melbourne School of Psychological Sciences The University of Melbourne Victoria Australia
- Orygen Research Centre Centre for Youth Mental Health University of Melbourne Victoria Australia
| |
Collapse
|
449
|
Egorov AI, Griffin SM, Converse RR, Styles JN, Sams EA, Wilson A, Jackson LE, Wade TJ. Vegetated land cover near residence is associated with reduced allostatic load and improved biomarkers of neuroendocrine, metabolic and immune functions. ENVIRONMENTAL RESEARCH 2017; 158:508-521. [PMID: 28709033 PMCID: PMC5941947 DOI: 10.1016/j.envres.2017.07.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/03/2017] [Accepted: 07/04/2017] [Indexed: 05/21/2023]
Abstract
BACKGROUND Greater exposure to urban green spaces has been linked to reduced risks of depression, cardiovascular disease, diabetes and premature death. Alleviation of chronic stress is a hypothesized pathway to improved health. Previous studies linked chronic stress with a biomarker-based composite measure of physiological dysregulation known as allostatic load. OBJECTIVE This study's objective was to assess the relationship between vegetated land cover near residences and allostatic load. METHODS This cross-sectional population-based study involved 206 adult residents of the Durham-Chapel Hill, North Carolina metropolitan area. Exposure was quantified using high-resolution metrics of trees and herbaceous vegetation within 500m of each residence derived from the U.S. Environmental Protection Agency's EnviroAtlas land cover dataset. Eighteen biomarkers of immune, neuroendocrine, and metabolic functions were measured in serum or saliva samples. Allostatic load was defined as a sum of potentially unhealthy biomarker values dichotomized at 10th or 90th percentile of sample distribution. Regression analysis was conducted using generalized additive models with two-dimensional spline smoothing function of geographic coordinates, weighted measures of vegetated land cover allowing decay of effects with distance, and geographic and demographic covariates. RESULTS An inter-quartile range increase in distance-weighted vegetated land cover was associated with 37% (95% Confidence Limits 46%; 27%) reduced allostatic load; significantly reduced adjusted odds of having low level of norepinephrine, dopamine, and dehydroepiandrosterone, and high level of epinephrine, fibrinogen, vascular cell adhesion molecule-1, and interleukin-8 in serum, and α-amylase in saliva; and reduced odds of previously diagnosed depression. CONCLUSIONS The observed effects of vegetated land cover on allostatic load and individual biomarkers are consistent with prevention of depression, cardiovascular disease and premature mortality.
Collapse
Affiliation(s)
- Andrey I Egorov
- National Health and Environmental Effects Research Laboratory, United States Environmental Protection Agency, Research Triangle Park, NC, USA.
| | - Shannon M Griffin
- National Exposure Research Laboratory, United States Environmental Protection Agency, Cincinnati, OH, USA
| | - Reagan R Converse
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jennifer N Styles
- National Health and Environmental Effects Research Laboratory, United States Environmental Protection Agency, Research Triangle Park, NC, USA; National Exposure Research Laboratory, United States Environmental Protection Agency, Cincinnati, OH, USA
| | - Elizabeth A Sams
- National Health and Environmental Effects Research Laboratory, United States Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Anthony Wilson
- Association of Schools and Programs of Public Health fellow at the United States Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Laura E Jackson
- National Health and Environmental Effects Research Laboratory, United States Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Timothy J Wade
- National Health and Environmental Effects Research Laboratory, United States Environmental Protection Agency, Research Triangle Park, NC, USA
| |
Collapse
|
450
|
Wu XM, Basu R, Malig B, Broadwin R, Ebisu K, Gold EB, Qi L, Derby C, Green RS. Association between gaseous air pollutants and inflammatory, hemostatic and lipid markers in a cohort of midlife women. ENVIRONMENT INTERNATIONAL 2017; 107:131-139. [PMID: 28732305 PMCID: PMC5584622 DOI: 10.1016/j.envint.2017.07.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 05/04/2023]
Abstract
BACKGROUND Exposures to ambient gaseous pollutants have been linked to cardiovascular diseases (CVDs), but the biological mechanisms remain uncertain. OBJECTIVES This study examined the changes in CVD marker levels resulting from elevated exposure to ambient gaseous pollutants in midlife women. METHODS Annual repeated measurements of several inflammatory, hemostatic and lipid makers were obtained from 2306 midlife women enrolled in the longitudinal Study of Women's Health Across the Nation (SWAN) between 1999 and 2004. Ambient carbon monoxide (CO), nitrogen dioxide (NO2), and sulfur dioxide (SO2) data were assigned to each woman based on proximity of the monitoring station to her residential address. Short- and long-term exposures were calculated, and their associations with markers were examined using linear mixed-effects regression models, adjusted for demographic, health and other factors. RESULTS Short-term CO exposure was associated with increased fibrinogen, i.e., every interquartile increase of average prior one-week exposure to CO was associated with 1.3% (95% CI: 0.6%, 2.0%) increase in fibrinogen. Long-term exposures to NO2 and SO2 were associated with reduced high-density lipoproteins and apolipoprotein A1, e.g., 4.0% (1.7%, 6.3%) and 4.7% (2.8%, 6.6%) decrease per interquartile increment in prior one-year average NO2 concentration, respectively. Fine particle (PM2.5) exposure confounded associations between CO/NO2 and inflammatory/hemostatic markers, while associations with lipoproteins were generally robust to PM2.5 adjustment. CONCLUSIONS Exposures to these gas pollutants at current ambient levels may increase thrombotic potential and disrupt cholesterol metabolism, contributing to greater risk of CVDs in midlife women. Caution should be exercised in evaluating the confounding by PM2.5 exposure.
Collapse
Affiliation(s)
- Xiangmei May Wu
- Air and Climate Epidemiology Section, Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, Oakland, CA, USA.
| | - Rupa Basu
- Air and Climate Epidemiology Section, Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, Oakland, CA, USA
| | - Brian Malig
- Air and Climate Epidemiology Section, Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, Oakland, CA, USA
| | - Rachel Broadwin
- Air and Climate Epidemiology Section, Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, Oakland, CA, USA
| | - Keita Ebisu
- Air and Climate Epidemiology Section, Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, Oakland, CA, USA
| | - Ellen B Gold
- Department of Public Health Sciences, University of California Davis, School of Medicine, Davis, CA, USA
| | - Lihong Qi
- Department of Public Health Sciences, University of California Davis, School of Medicine, Davis, CA, USA
| | - Carol Derby
- Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rochelle S Green
- Air and Climate Epidemiology Section, Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, Oakland, CA, USA
| |
Collapse
|