651
|
Abstract
The importance of total cardiovascular (CV) risk estimation before management decisions are taken is well established. Models have been developed that allow physicians to stratify the asymptomatic population in subgroups at low, moderate, high, and very high total CV risk. Most models are based on classical CV risk factors: age, gender, smoking, blood pressure, and lipid levels. The impact of additional risk factors is discussed here, looking separately at the predictive increments of novel biomarkers and of indicators of subclinical atherosclerotic disease. The contribution of biomarkers to the total CV risk estimation is generally modest, and their usage should be limited to subjects at intermediate total CV risk. Detection of subclinical vascular damage may improve total CV risk estimation in asymptomatic subjects who are close to a threshold that could affect management decisions and in whom the chances of re-classification in a different risk category are great. There is, however, an urgent need for trials in which the value of using total CV risk estimation models is tested.
Collapse
|
652
|
Wang X, Hunter D, Xu J, Ding C. Metabolic triggered inflammation in osteoarthritis. Osteoarthritis Cartilage 2015; 23:22-30. [PMID: 25452156 DOI: 10.1016/j.joca.2014.10.002] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 09/23/2014] [Accepted: 10/06/2014] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is a common chronic joint disorder with a multifactorial etiology including genetic and environmental factors. Metabolic triggered inflammation, induced by nutrient overload and metabolic surplus, consists of components such as obesity, pro-inflammatory cytokines and adipokines, abnormal metabolites, acute phase proteins, vitamin D deficiency, and deregulated microRNAs that may play a role in OA pathophysiology. Obesity-related metabolic factors, especially adipokines, contribute to OA development by inducing pro-inflammatory cytokines and degradative enzymes, leading to cartilage matrix impairment and subchondral bone remodeling. Ectopic metabolite deposition and low-grade systemic inflammation can contribute to a toxic internal environment that exacerbates OA. Complement components highly expressed in osteoarthritic joints have also been proposed as causative factors. Vitamin D deficiency has been associated with obesity and is implicated to be associated with cartilage loss in OA. Metabolic microRNAs may explain the inflammatory link between obesity and OA. Therapies targeting metabolic-triggered inflammation and its components are anticipated to have potential for the treatment of OA.
Collapse
Affiliation(s)
- X Wang
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania, Australia
| | - D Hunter
- Institute of Bone and Joint Research, Kolling Institute and Royal North Shore Hospital, University of Sydney, Sydney, NSW, Australia
| | - J Xu
- Department of Rheumatology and Arthritis Research Institute, 1st Affiliated Hospital, Anhui Medical University, Hefei, China
| | - C Ding
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania, Australia; Institute of Bone and Joint Research, Kolling Institute and Royal North Shore Hospital, University of Sydney, Sydney, NSW, Australia; Department of Rheumatology and Arthritis Research Institute, 1st Affiliated Hospital, Anhui Medical University, Hefei, China.
| |
Collapse
|
653
|
Association of β-fibrinogen promoter gene polymorphism (−148C/T), hyperfibrinogenemia and ischemic stroke in young adult patients. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2015. [DOI: 10.1016/j.ejmhg.2014.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
654
|
Kyjovska ZO, Jacobsen NR, Saber AT, Bengtson S, Jackson P, Wallin H, Vogel U. DNA damage following pulmonary exposure by instillation to low doses of carbon black (Printex 90) nanoparticles in mice. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2015; 56:41-9. [PMID: 25042074 PMCID: PMC4312987 DOI: 10.1002/em.21888] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 07/04/2014] [Indexed: 05/27/2023]
Abstract
We previously observed genotoxic effects of carbon black nanoparticles at low doses relative to the Danish Occupational Exposure Limit (3.5 mg/m(3)). Furthermore, DNA damage occurred in broncho-alveolar lavage (BAL) cells in the absence of inflammation, indicating that inflammation is not required for the genotoxic effects of carbon black. In this study, we investigated inflammatory and acute phase response in addition to genotoxic effects occurring following exposure to nanoparticulate carbon black (NPCB) at even lower doses. C57BL/6JBomTac mice were examined 1, 3, and 28 days after a single instillation of 0.67, 2, 6, and 162 µg Printex 90 NPCB and vehicle. Cellular composition and protein concentration was evaluated in BAL fluid as markers of inflammatory response and cell damage. DNA strand breaks in BAL cells, lung, and liver tissue were assessed using the alkaline comet assay. The pulmonary acute phase response was analyzed by Saa3 mRNA real-time quantitative PCR. Instillation of the low doses of NPCB induced a slight neutrophil influx one day after exposure. Pulmonary exposure to small doses of NPCB caused an increase in DNA strand breaks in BAL cells and lung tissue measured using the comet assay. We interpret the increased DNA strand breaks occurring following these low exposure doses of NPCB as DNA damage caused by primary genotoxicity in the absence of substantial inflammation, cell damage, and acute phase response.
Collapse
Affiliation(s)
- Zdenka O Kyjovska
- Danish Centre for Nanosafety, National Research Centre for the Working EnvironmentCopenhagen Ø, Denmark
| | - Nicklas R Jacobsen
- Danish Centre for Nanosafety, National Research Centre for the Working EnvironmentCopenhagen Ø, Denmark
| | - Anne T Saber
- Danish Centre for Nanosafety, National Research Centre for the Working EnvironmentCopenhagen Ø, Denmark
| | - Stefan Bengtson
- Danish Centre for Nanosafety, National Research Centre for the Working EnvironmentCopenhagen Ø, Denmark
| | - Petra Jackson
- Danish Centre for Nanosafety, National Research Centre for the Working EnvironmentCopenhagen Ø, Denmark
| | - Håkan Wallin
- Danish Centre for Nanosafety, National Research Centre for the Working EnvironmentCopenhagen Ø, Denmark
| | - Ulla Vogel
- Danish Centre for Nanosafety, National Research Centre for the Working EnvironmentCopenhagen Ø, Denmark
- Department of Micro- and Nanotechnology, Technical University of DenmarkLyngby, Denmark
| |
Collapse
|
655
|
Racial disparities in the health benefits of educational attainment: a study of inflammatory trajectories among African American and white adults. Psychosom Med 2015; 77:33-40. [PMID: 25490696 DOI: 10.1097/psy.0000000000000128] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE The current study examined the prospective effects of educational attainment on proinflammatory physiology among African American and white adults. METHODS Participants were 1192 African Americans and 1487 whites who participated in Year 5 (mean [standard deviation] age = 30 [3.5] years), and Year 20 (mean [standard deviation] age = 45 [3.5]) of an ongoing longitudinal study. Initial analyses focused on age-related changes in fibrinogen across racial groups, and parallel analyses for C-reactive protein and interleukin-6 assessed at Year 20. Models then estimated the effects of educational attainment on changes in inflammation for African Americans and whites before and after controlling for four blocks of covariates: a) early life adversity, b) health and health behaviors at baseline, c) employment and financial measures at baseline and follow-up, and d) psychosocial stresses in adulthood. RESULTS African Americans had larger increases in fibrinogen over time than whites (B = 24.93, standard error = 3.24, p < .001), and 37% of this difference was explained after including all covariates. Effects of educational attainment were weaker for African Americans than for whites (B = 10.11, standard error = 3.29, p = .002), and only 8% of this difference was explained by covariates. Analyses for C-reactive protein and interleukin-6 yielded consistent results. CONCLUSIONS The effects of educational attainment on inflammation levels were stronger for white than for African American participants. Why African Americans do not show the same health benefits with educational attainment is an important question for health disparities research.
Collapse
|
656
|
Paepegaey AC, Genser L, Bouillot JL, Oppert JM, Clément K, Poitou C. High levels of CRP in morbid obesity: the central role of adipose tissue and lessons for clinical practice before and after bariatric surgery. Surg Obes Relat Dis 2015; 11:148-54. [DOI: 10.1016/j.soard.2014.06.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 06/09/2014] [Accepted: 06/10/2014] [Indexed: 01/26/2023]
|
657
|
Turgeon RD, Anderson TJ, Grégoire J, Pearson GJ. Updated guidelines for the management of dyslipidemia and prevention of cardiovascular disease by pharmacists. Can Pharm J (Ott) 2015; 148:21-8. [PMID: 26759562 PMCID: PMC4294807 DOI: 10.1177/1715163514561256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Ricky D Turgeon
- Faculty of Pharmaceutical Sciences (Turgeon), University of British Columbia, Vancouver, BC
| | - Todd J Anderson
- Faculty of Pharmaceutical Sciences (Turgeon), University of British Columbia, Vancouver, BC
| | - Jean Grégoire
- Faculty of Pharmaceutical Sciences (Turgeon), University of British Columbia, Vancouver, BC
| | - Glen J Pearson
- Faculty of Pharmaceutical Sciences (Turgeon), University of British Columbia, Vancouver, BC
| |
Collapse
|
658
|
High sensitive C-reactive protein (hsCRP), cardiovascular events and mortality in the aged: A prospective 9-year follow-up study. Arch Gerontol Geriatr 2015; 60:112-7. [DOI: 10.1016/j.archger.2014.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 03/10/2014] [Accepted: 10/06/2014] [Indexed: 11/20/2022]
|
659
|
Baumert J, Huang J, McKnight B, Sabater-Lleal M, Steri M, Chu AY, Trompet S, Lopez LM, Fornage M, Teumer A, Tang W, Rudnicka AR, Mälarstig A, Hottenga JJ, Kavousi M, Lahti J, Tanaka T, Hayward C, Huffman JE, Morange PE, Rose LM, Basu S, Rumley A, Stott DJ, Buckley BM, de Craen AJM, Sanna S, Masala M, Biffar R, Homuth G, Silveira A, Sennblad B, Goel A, Watkins H, Müller-Nurasyid M, Rückerl R, Taylor K, Chen MH, de Geus EJC, Hofman A, Witteman JCM, de Maat MPM, Palotie A, Davies G, Siscovick DS, Kolcic I, Wild SH, Song J, McArdle WL, Ford I, Sattar N, Schlessinger D, Grotevendt A, Franzosi MG, Illig T, Waldenberger M, Lumley T, Tofler GH, Willemsen G, Uitterlinden AG, Rivadeneira F, Räikkönen K, Chasman DI, Folsom AR, Lowe GD, Westendorp RGJ, Slagboom PE, Cucca F, Wallaschofski H, Strawbridge RJ, Seedorf U, Koenig W, Bis JC, Mukamal KJ, van Dongen J, Widen E, Franco OH, Starr JM, Liu K, Ferrucci L, Polasek O, Wilson JF, Oudot-Mellakh T, Campbell H, Navarro P, Bandinelli S, Eriksson J, Boomsma DI, Dehghan A, Clarke R, Hamsten A, Boerwinkle E, Jukema JW, Naitza S, Ridker PM, Völzke H, Deary IJ, Reiner AP, Trégouët DA, O'Donnell CJ, et alBaumert J, Huang J, McKnight B, Sabater-Lleal M, Steri M, Chu AY, Trompet S, Lopez LM, Fornage M, Teumer A, Tang W, Rudnicka AR, Mälarstig A, Hottenga JJ, Kavousi M, Lahti J, Tanaka T, Hayward C, Huffman JE, Morange PE, Rose LM, Basu S, Rumley A, Stott DJ, Buckley BM, de Craen AJM, Sanna S, Masala M, Biffar R, Homuth G, Silveira A, Sennblad B, Goel A, Watkins H, Müller-Nurasyid M, Rückerl R, Taylor K, Chen MH, de Geus EJC, Hofman A, Witteman JCM, de Maat MPM, Palotie A, Davies G, Siscovick DS, Kolcic I, Wild SH, Song J, McArdle WL, Ford I, Sattar N, Schlessinger D, Grotevendt A, Franzosi MG, Illig T, Waldenberger M, Lumley T, Tofler GH, Willemsen G, Uitterlinden AG, Rivadeneira F, Räikkönen K, Chasman DI, Folsom AR, Lowe GD, Westendorp RGJ, Slagboom PE, Cucca F, Wallaschofski H, Strawbridge RJ, Seedorf U, Koenig W, Bis JC, Mukamal KJ, van Dongen J, Widen E, Franco OH, Starr JM, Liu K, Ferrucci L, Polasek O, Wilson JF, Oudot-Mellakh T, Campbell H, Navarro P, Bandinelli S, Eriksson J, Boomsma DI, Dehghan A, Clarke R, Hamsten A, Boerwinkle E, Jukema JW, Naitza S, Ridker PM, Völzke H, Deary IJ, Reiner AP, Trégouët DA, O'Donnell CJ, Strachan DP, Peters A, Smith NL. No evidence for genome-wide interactions on plasma fibrinogen by smoking, alcohol consumption and body mass index: results from meta-analyses of 80,607 subjects. PLoS One 2014; 9:e111156. [PMID: 25551457 PMCID: PMC4281156 DOI: 10.1371/journal.pone.0111156] [Show More Authors] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 09/23/2014] [Indexed: 11/23/2022] Open
Abstract
Plasma fibrinogen is an acute phase protein playing an important role in the blood coagulation cascade having strong associations with smoking, alcohol consumption and body mass index (BMI). Genome-wide association studies (GWAS) have identified a variety of gene regions associated with elevated plasma fibrinogen concentrations. However, little is yet known about how associations between environmental factors and fibrinogen might be modified by genetic variation. Therefore, we conducted large-scale meta-analyses of genome-wide interaction studies to identify possible interactions of genetic variants and smoking status, alcohol consumption or BMI on fibrinogen concentration. The present study included 80,607 subjects of European ancestry from 22 studies. Genome-wide interaction analyses were performed separately in each study for about 2.6 million single nucleotide polymorphisms (SNPs) across the 22 autosomal chromosomes. For each SNP and risk factor, we performed a linear regression under an additive genetic model including an interaction term between SNP and risk factor. Interaction estimates were meta-analysed using a fixed-effects model. No genome-wide significant interaction with smoking status, alcohol consumption or BMI was observed in the meta-analyses. The most suggestive interaction was found for smoking and rs10519203, located in the LOC123688 region on chromosome 15, with a p value of 6.2×10−8. This large genome-wide interaction study including 80,607 participants found no strong evidence of interaction between genetic variants and smoking status, alcohol consumption or BMI on fibrinogen concentrations. Further studies are needed to yield deeper insight in the interplay between environmental factors and gene variants on the regulation of fibrinogen concentrations.
Collapse
Affiliation(s)
- Jens Baumert
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Jie Huang
- National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, United States of America
- National Heart, Lung and Blood Institute Division of Intramural Research, Bethesda, Maryland, United States of America
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Barbara McKnight
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Maria Sabater-Lleal
- Cardiovascular Genetics and Genomics Group, Atherosclerosis Research Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Maristella Steri
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cagliari, Italy
| | - Audrey Y. Chu
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Lorna M. Lopez
- Department of Psychology, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, Division of Epidemiology, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Alexander Teumer
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Weihong Tang
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Alicja R. Rudnicka
- Division of Population Health Sciences & Education, St George's, University of London, Cranmer Terrace, London, United Kingdom
| | - Anders Mälarstig
- Cardiovascular Genetics and Genomics Group, Atherosclerosis Research Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, VU University & EMGO+ institute, VU Medical Centre, Amsterdam, the Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Netherlands Genomics Initiative (NGI)-Sponsored Netherlands Consortium for Healthy Aging (NCHA), Rotterdam, the Netherlands
| | - Jari Lahti
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland
- Folkhalsan Research Centre, Helsinki, Finland
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, Scotland, United Kingdom
| | - Jennifer E. Huffman
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, Scotland, United Kingdom
| | | | - Lynda M. Rose
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Saonli Basu
- Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ann Rumley
- Division of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - David J. Stott
- Institute of Cardiovascular and Medical Sciences, Faculty of Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Brendan M. Buckley
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Anton J. M. de Craen
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Serena Sanna
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cagliari, Italy
| | - Marco Masala
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cagliari, Italy
| | - Reiner Biffar
- Department of Prosthetic Dentistry, Gerostomatology and Dental Materials, University Medicine Greifswald, Greifswald, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Angela Silveira
- Cardiovascular Genetics and Genomics Group, Atherosclerosis Research Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Bengt Sennblad
- Cardiovascular Genetics and Genomics Group, Atherosclerosis Research Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
- Science for Life Laboratory, Karolinska Insitutet, Stockholm, Sweden
| | - Anuj Goel
- Department of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
- Department of Cardiovascular Medicine, The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Hugh Watkins
- Department of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
- Department of Cardiovascular Medicine, The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Regina Rückerl
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- ESC-Environmental Science Center, University of Augsburg, Augsburg, Germany
| | - Kent Taylor
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Ming-Huei Chen
- Department of Biostatistics, Boston University, Boston, Massachusetts, United States of America
| | - Eco J. C. de Geus
- Department of Haematology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Netherlands Genomics Initiative (NGI)-Sponsored Netherlands Consortium for Healthy Aging (NCHA), Rotterdam, the Netherlands
| | - Jacqueline C. M. Witteman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Netherlands Genomics Initiative (NGI)-Sponsored Netherlands Consortium for Healthy Aging (NCHA), Rotterdam, the Netherlands
| | | | - Aarno Palotie
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, Finland
| | - Gail Davies
- Department of Psychology, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - David S. Siscovick
- Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Ivana Kolcic
- Department of Public Health, University of Split Medical School, Split, Croatia
| | - Sarah H. Wild
- Centre for Population Health Sciences, University of Edinburgh, Teviot Place, Edinburgh, Scotland, United Kingdom
| | - Jaejoon Song
- Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Wendy L. McArdle
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Ian Ford
- Robertson Center for Biostatistics, University of Glasgow, Glasgow, United Kingdom
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, Faculty of Medicine, Glasgow, United Kingdom
| | - David Schlessinger
- Intramural Research Program, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Anne Grotevendt
- Institute for Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Maria Grazia Franzosi
- Department of Cardiovascular Research, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Lumley
- Department of Statistics, University of Auckland, Auckland, New Zealand
| | | | - Gonneke Willemsen
- Department of Biological Psychology, VU University & EMGO+ institute, VU Medical Centre, Amsterdam, the Netherlands
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Netherlands Genomics Initiative (NGI)-Sponsored Netherlands Consortium for Healthy Aging (NCHA), Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Fernando Rivadeneira
- Division of Preventive Medicine, Division of Genetics, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Katri Räikkönen
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland
| | - Daniel I. Chasman
- Division of Preventive Medicine, Division of Genetics, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Aaron R. Folsom
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Gordon D. Lowe
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Rudi G. J. Westendorp
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - P. Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cagliari, Italy
| | - Henri Wallaschofski
- Intramural Research Program, National Institute on Aging, Baltimore, Maryland, United States of America
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Rona J. Strawbridge
- Cardiovascular Genetics and Genomics Group, Atherosclerosis Research Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Udo Seedorf
- Leibniz-Institut für Arterioskleroseforschung an der Universität Münster, Münster, Germany
| | - Wolfgang Koenig
- Department of Internal Medicine II - Cardiology, University of Ulm Medical Center, Ulm, Germany
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Kenneth J. Mukamal
- Harvard Medical School, Boston, Massachusetts, United States of America
- Division of General Medicine and Primary Care, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Jenny van Dongen
- Department of Biological Psychology, VU University & EMGO+ institute, VU Medical Centre, Amsterdam, the Netherlands
| | - Elisabeth Widen
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - Oscar H. Franco
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Netherlands Genomics Initiative (NGI)-Sponsored Netherlands Consortium for Healthy Aging (NCHA), Rotterdam, the Netherlands
| | - John M. Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
- Alzheimer Scotland Dementia Research Centre, The University of Edinburgh, Edinburgh, United Kingdom
| | - Kiang Liu
- Department of Preventive Medicine, Northwestern University Medical School, Chicago, Illinois, United States of America
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Ozren Polasek
- Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - James F. Wilson
- Department of Public Health, University of Split Medical School, Split, Croatia
| | - Tiphaine Oudot-Mellakh
- INSERM, UMR_S 1166, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1166, Team Genomics & Pathophysiology of Cardiovascular Diseases, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Harry Campbell
- Department of Public Health, University of Split Medical School, Split, Croatia
| | - Pau Navarro
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, Scotland, United Kingdom
| | | | - Johan Eriksson
- Folkhalsan Research Centre, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
- Helsinki University Central Hospital, Unit of General Practice, Helsinki, Finland
| | - Dorret I. Boomsma
- Department of Biological Psychology, VU University & EMGO+ institute, VU Medical Centre, Amsterdam, the Netherlands
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Netherlands Genomics Initiative (NGI)-Sponsored Netherlands Consortium for Healthy Aging (NCHA), Rotterdam, the Netherlands
| | - Robert Clarke
- Clinical Trial Service Unit, University of Oxford, Oxford, United Kingdom
| | - Anders Hamsten
- Cardiovascular Genetics and Genomics Group, Atherosclerosis Research Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Eric Boerwinkle
- Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, United States of America
| | - J. Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Durrer Center for Cardiogenetic Research, Amsterdam, the Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, the Netherlands
| | - Silvia Naitza
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cagliari, Italy
| | - Paul M. Ridker
- Division of Preventive Medicine, Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Ian J. Deary
- Department of Psychology, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Alexander P. Reiner
- Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, Finland
| | - David-Alexandre Trégouët
- INSERM, UMR_S 1166, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1166, Team Genomics & Pathophysiology of Cardiovascular Diseases, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Christopher J. O'Donnell
- National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, United States of America
- National Heart, Lung and Blood Institute Division of Intramural Research, Bethesda, Maryland, United States of America
| | - David P. Strachan
- Division of Population Health Sciences & Education, St George's, University of London, Cranmer Terrace, London, United Kingdom
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich, Munich, Germany
- * E-mail: (A. Peters); (NLS)
| | - Nicholas L. Smith
- Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, Finland
- Group Health Research Institute, Group Health Cooperative, Seattle, Washington, United States of America
- Seattle Epidemiologic Research & Information Center, Veterans Affairs Office of Research & Development, Seattle, Washington, United States of America
- * E-mail: (A. Peters); (NLS)
| |
Collapse
|
660
|
Bian F, Yang X, Zhou F, Wu PH, Xing S, Xu G, Li W, Chi J, Ouyang C, Zhang Y, Xiong B, Li Y, Zheng T, Wu D, Chen X, Jin S. C-reactive protein promotes atherosclerosis by increasing LDL transcytosis across endothelial cells. Br J Pharmacol 2014; 171:2671-84. [PMID: 24517733 DOI: 10.1111/bph.12616] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 09/30/2013] [Accepted: 10/01/2013] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE The retention of plasma low-density lipoprotein (LDL) particles in subendothelial space following transcytosis across the endothelium is the initial step of atherosclerosis. Whether or not C-reactive protein (CRP) can directly affect the transcytosis of LDL is not clear. Here we have examined the effect of CRP on transcytosis of LDL across endothelial cells and have explored the underlying mechanisms. EXPERIMENTAL APPROACH Effects of CRP on transcytosis of FITC-labelled LDL were examined with human umbilical vein endothelial cells and venous rings in vitro and, in vivo, ApoE(-/-) mice. Laser scanning confocal microscopy, immunohistochemistry and Oil Red O staining were used to assay LDL. KEY RESULTS CRP increased transcytosis of LDL. An NADPH oxidase inhibitor, diphenylene iodonium, and the reducing agent, dithiothreitol partly or completely blocked CRP-stimulated increase of LDL transcytosis. The PKC inhibitor, bisindolylmaleimide I and the Src kinase inhibitor, PP2, blocked the trafficking of the molecules responsible for transcytosis. Confocal imaging analysis revealed that CRP stimulated LDL uptake by endothelial cells and vessel walls. In ApoE(-/-) mice, CRP significantly promoted early changes of atherosclerosis, which were blocked by inhibitors of transcytosis. CONCLUSIONS AND IMPLICATIONS CRP promoted atherosclerosis by directly increasing the transcytosis of LDL across endothelial cells and increasing LDL retention in vascular walls. These actions of CRP were associated with generation of reactive oxygen species, activation of PKC and Src, and translocation of caveolar or soluble forms of the N-ethylmaleimide-sensitive factor attachment protein.
Collapse
Affiliation(s)
- Fang Bian
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, The Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
661
|
Leite WF, Ramires JAF, Moreira LFP, Strunz CMC, Mangione JA. Correlation between C-reactive protein in peripheral vein and coronary sinus in stable and unstable angina. Arq Bras Cardiol 2014; 104:202-8. [PMID: 25494014 PMCID: PMC4386848 DOI: 10.5935/abc.20140188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 09/30/2014] [Indexed: 01/18/2023] Open
Abstract
Background High sensitivity C-reactive protein (hs-CRP) is commonly used in clinical practice
to assess cardiovascular risk. However, a correlation has not yet been established
between the absolute levels of peripheral and central hs-CRP. Objective To assess the correlation between serum hs-CRP levels (mg/L) in a peripheral vein
in the left forearm (LFPV) with those in the coronary sinus (CS) of patients with
coronary artery disease (CAD) and a diagnosis of stable angina (SA) or unstable
angina (UA). Methods This observational, descriptive, and cross-sectional study was conducted at the
Instituto do Coração, Hospital das Clinicas, Faculdade de Medicina, Universidade
de São Paulo, and at the Hospital Beneficência Portuguesa de Sao Paulo, where CAD
patients referred to the hospital for coronary angiography were evaluated. Results Forty patients with CAD (20 with SA and 20 with UA) were included in the study.
Blood samples from LFPV and CS were collected before coronary angiography.
Furthermore, analysis of the correlation between serum levels of hs-CRP in LFPV
versus CS showed a strong linear correlation for both SA (r = 0.993, p < 0.001)
and UA (r = 0.976, p < 0.001) and for the entire sample (r = 0.985, p <
0.001). Conclusion Our data suggest a strong linear correlation between hs-CRP levels in LFPV versus
CS in patients with SA and UA.
Collapse
Affiliation(s)
- Weverton Ferreira Leite
- Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | | | | |
Collapse
|
662
|
Luchetti M, Barkley JM, Stephan Y, Terracciano A, Sutin AR. Five-factor model personality traits and inflammatory markers: new data and a meta-analysis. Psychoneuroendocrinology 2014; 50:181-93. [PMID: 25233337 PMCID: PMC4544833 DOI: 10.1016/j.psyneuen.2014.08.014] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 08/18/2014] [Accepted: 08/25/2014] [Indexed: 01/25/2023]
Abstract
The purpose of this research is to examine the association between five major dimensions of personality and systemic inflammation through (a) new data on C-reactive protein (CRP) from three large national samples of adults that together cover most of the adult lifespan and (b) a meta-analysis of published studies on CRP and interleukin-6 (IL-6). New data (total N=26,305) were drawn from the National Longitudinal Study of Adolescent Health, the Midlife in the United States study, and the Health and Retirement Study. PRISMA guidelines were used for the meta-analysis to combine results of up to seven studies on CRP (N=34,067) and six on IL-6 (N=7538). Across the three new samples, higher conscientiousness was associated with lower CRP. The conscientiousness-CRP relation was virtually identical controlling for smoking; controlling for body mass index attenuated this association but did not eliminate it. Compared to participants in the highest quartile of conscientiousness, participants in the lowest quartile had an up to 50% increased risk of CRP levels that exceeded the clinical threshold (≥3 mg/l). The meta-analysis supported the association between conscientiousness and both CRP and IL-6 and also suggested a negative association between openness and CRP; no associations were found for neuroticism, extraversion and agreeableness. The present work indicates a modest, but consistent, association between conscientiousness and a more favorable inflammatory profile, which may contribute to the role of conscientiousness in better health across the lifespan.
Collapse
Affiliation(s)
- Martina Luchetti
- Department of Psychology University of Bologna, Viale Berti Pichat 5, Bologna 40127, Italy.
| | - James M. Barkley
- Tallahassee Community College, 444 Appleyard Drive, Tallahassee, FL 32304,
| | - Yannick Stephan
- EA 4556 EPSYLON, Laboratory Dynamic of Human Abilities and Health Behaviors, Department of Sport Sciences, Psychology and Medicine, University of Montpellier and St Etienne, 4, Boulevard Henry IV, 34000 Montpellier, France.
| | - Antonio Terracciano
- Department of Geriatrics, Florida State University College of Medicine, 1115 W. Call Street, Tallahassee, FL 32306, USA.
| | - Angelina R. Sutin
- Department of Behavioral Sciences and Social Medicine, Florida State University College of Medicine, 1115 W. Call Street, Tallahassee, FL, 32306, Phone: +1 850 645 0438,
| |
Collapse
|
663
|
Insights from a thermography-based method suggesting higher carotid inflammation in patients with diabetes mellitus and coronary artery disease. DIABETES & METABOLISM 2014; 40:431-8. [DOI: 10.1016/j.diabet.2014.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 05/08/2014] [Accepted: 05/13/2014] [Indexed: 12/30/2022]
|
664
|
Sesti G, Fiorentino TV, Succurro E, Perticone M, Arturi F, Sciacqua A, Perticone F. Elevated 1-h post-load plasma glucose levels in subjects with normal glucose tolerance are associated with unfavorable inflammatory profile. Acta Diabetol 2014; 51:927-32. [PMID: 24619654 DOI: 10.1007/s00592-013-0539-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 11/28/2013] [Indexed: 02/06/2023]
Abstract
Glucose tolerant subjects with 1-h post-load glucose ≥155 mg/dl (NGT-1 h-high) are at increased risk for type 2 diabetes (T2DM). Prospective studies showed that chronic subclinical inflammation is a predictor of T2DM. In this study, we aimed to evaluate the inflammatory profile in NGT-1 h-high subjects as compared with individuals with 1-h post-load glucose <155 mg/dl (NGT-1 h-low). To this end, an oral glucose tolerance tests (OGTT) were performed in 1,099 nondiabetic whites. Cardio-metabolic risk factors including high-sensitivity C-reactive protein (hsCRP), erythrocyte sedimentation rate (ESR), fibrinogen, and complement C3 (C3) were determined. Of the 1,099 subjects examined, 497 had NGT-1 h-low, 154 had NGT-1 h-high, 158 had isolated impaired fasting glucose (IFG), and 290 had impaired glucose tolerance (IGT). As compared with NGT-1 h-low, NGT-1 h-high and IGT subjects exhibited significantly higher hsCRP, ESR, fibrinogen, and C3 levels. Notably, hsCRP, ESR, and C3 were also significantly higher as compared with IFG individuals. In a logistic regression analysis adjusted for age and gender, NGT-1 h-high and IGT subjects had a 1.8-fold increased risk of having the highest value of the Inflammatory Score. These data suggest that a value of a 1-h OGTT glucose ≥155 mg/dl may be helpful to identify a subset of normal glucose tolerance individuals at risk for chronic subclinical inflammation, a predictor of T2DM, and cardiovascular diseases.
Collapse
Affiliation(s)
- Giorgio Sesti
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, 88100, Viale Europa, Catanzaro, Italy,
| | | | | | | | | | | | | |
Collapse
|
665
|
|
666
|
Price AH, Welsh P, Weir CJ, Feinkohl I, Robertson CM, Morling JR, McLachlan S, Strachan MWJ, Sattar N, Price JF. N-terminal pro-brain natriuretic peptide and risk of cardiovascular events in older patients with type 2 diabetes: the Edinburgh Type 2 Diabetes Study. Diabetologia 2014; 57:2505-12. [PMID: 25231020 DOI: 10.1007/s00125-014-3375-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 08/26/2014] [Indexed: 12/22/2022]
Abstract
AIMS/HYPOTHESIS The aim of this study was to investigate the association of N-terminal pro-brain natriuretic peptide (NT-proBNP) with traditional cardiovascular risk factors and incident cardiovascular events in older people with type 2 diabetes. METHODS In the prospective phase of the Edinburgh Type 2 Diabetes Study, 1066 men and women aged 60 to 75 years with type 2 diabetes mellitus were followed for 4 years; 112 participants had an incident cardiovascular event. At baseline, cardiovascular risk factors, pre-existing cardiovascular disease and levels of NT-proBNP were evaluated. RESULTS Raised plasma NT-proBNP levels were associated with these classical cardiovascular risk factors: increased duration of diabetes, use of insulin, raised BMI, reduced HDL-cholesterol, reduced renal function and use of lipid-lowering and anti-hypertensive medication (all p < 0.05). In the prospective analysis, NT-proBNP was strongly associated with subsequent risk of all cardiovascular disease events (HR per one SD increase in NT-proBNP 1.39; 95% CI 1.10, 1.75), independent of cardiovascular risk factors traditionally used to predict vascular events. NT-proBNP was also independently associated with incident coronary artery disease events (1.48, 95% CI 1.10, 1.98). The addition of NT-proBNP to multivariate models improved the C-index by 0.019 for the 'hard' cardiac endpoint (fatal and non-fatal myocardial infarction). CONCLUSIONS/INTERPRETATION In older people with type 2 diabetes, NT-proBNP is associated with the development of coronary and cerebrovascular events, independent of a wide range of other vascular and metabolic risk factors, and may prove a useful addition to current vascular risk scores in diabetes populations.
Collapse
Affiliation(s)
- Anna H Price
- Centre for Population Health Sciences, The University of Edinburgh Medical School, Teviot Place, Edinburgh, Scotland, EH8 9AG, UK,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
667
|
Moosheer SM, Waldschütz W, Itariu BK, Brath H, Stulnig TM. A protein-enriched low glycemic index diet with omega-3 polyunsaturated fatty acid supplementation exerts beneficial effects on metabolic control in type 2 diabetes. Prim Care Diabetes 2014; 8:308-314. [PMID: 24656509 DOI: 10.1016/j.pcd.2014.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 02/19/2014] [Accepted: 02/24/2014] [Indexed: 11/20/2022]
Abstract
AIMS The current study aims to investigate practicability and effects of a combined dietary intervention with increased relative protein content supplemented with omega-3 polyunsaturated fatty acids (PUFA) on metabolic control and inflammatory parameters in a real life situation in type 2 diabetes patients. METHODS In this observational study we advised thirty mostly obese patients with type 2 diabetes to follow a protein-enriched diet with carbohydrates of low glycemic index (low GI) and moderate fat reduction supplemented with omega-3 PUFA for 24 weeks. Primary efficacy parameter was the change in HbA1c; secondary parameters included changes in systemic inflammation (measured by ultrasensitive C-reactive protein, usCRP), body weight, waist circumference, fat mass. The study is registered at clinicaltrials.gov (NCT01474603). RESULTS The dietary intervention significantly reduced the primary efficacy variable HbA1c from a baseline value of 63±11mmol/mol to 59±14mmol/mol (P=0.033) and 56±12mmol/mol (P=0.001) after 12 and 24 weeks, respectively. In addition, usCRP decreased significantly at 24 weeks (P=0.039). Waist circumference, an important indicator for cardiometabolic-risk and silent inflammation, decreased from baseline 116.0±14.1cm to 114.9±13.5cm (P=0.019), 114.0±14.4cm (P=0.001), and 112.7±13.4cm (P=0.049), after 3, 12 and 24 weeks, respectively. CONCLUSION Counseling a protein enriched and low glycemic index diet supplemented with long-chain omega-3 PUFA in a real-life clinical setting improves glycemic control and also reduces waist circumference and silent inflammation in overweight or obese patients with type 2 diabetes.
Collapse
Affiliation(s)
- Simone M Moosheer
- Christian Doppler Laboratory for Cardio-Metabolic Immunotherapy and Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Austria
| | | | - Bianca K Itariu
- Christian Doppler Laboratory for Cardio-Metabolic Immunotherapy and Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Austria
| | - Helmut Brath
- Diabetes Outpatient Clinic, Health Center South, Vienna, Austria
| | - Thomas M Stulnig
- Christian Doppler Laboratory for Cardio-Metabolic Immunotherapy and Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Austria.
| |
Collapse
|
668
|
Sun C, Magnussen CG, Ponsonby AL, Schmidt MD, Carlin JB, Huynh Q, Venn AJ, Dwyer T. The contribution of childhood cardiorespiratory fitness and adiposity to inflammation in young adults. Obesity (Silver Spring) 2014; 22:2598-605. [PMID: 25297830 DOI: 10.1002/oby.20871] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 07/23/2014] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Cardiorespiratory fitness and adiposity may influence cardiovascular risk through their effects on inflammation. The long-term effects of these modifiable factors on adult inflammation remain uncertain. The associations of childhood and adulthood cardiorespiratory fitness and adiposity with adult inflammation [C-reactive protein (CRP), fibrinogen] were examined. METHODS 1,976 children examined in 1985 and re-examined as young adults in 2004-2006 were included. Cardiorespiratory fitness and adiposity were assessed at both waves. CRP and fibrinogen were measured at follow-up. RESULTS Higher childhood fitness was associated with lower adult inflammation in both sexes. After adjusting for childhood adiposity, the association with CRP attenuated in males, but remained in females (average reduction of CRP 18.1% (95% CI 11.3-24.4%) per 1-SD increase in childhood fitness). Higher adult fitness, adjusting for childhood fitness (an increase in fitness from childhood to adulthood), was associated with lower adult CRP in females and lower fibrinogen in males. Higher childhood and adulthood adiposity (an increase in adiposity from childhood to adulthood) were associated with higher adult inflammation in both sexes. CONCLUSIONS Prevention programs to increase fitness and reduce adiposity in childhood, and maintain a favorable fitness and weight into adulthood, may lead to reduction in adult systemic inflammation.
Collapse
Affiliation(s)
- Cong Sun
- Environmental and Genetic Epidemiology Research Group, Murdoch Children's Research Institute (MCRI), Royal Children's Hospital, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | |
Collapse
|
669
|
Zhang Y, Zhu CG, Guo YL, Xu RX, Li S, Dong Q, Li JJ. Higher fibrinogen level is independently linked with the presence and severity of new-onset coronary atherosclerosis among Han Chinese population. PLoS One 2014; 9:e113460. [PMID: 25426943 PMCID: PMC4245131 DOI: 10.1371/journal.pone.0113460] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 10/26/2014] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Fibrinogen is a coagulation/inflammatory biomarker strongly associated with atherogenesis. However, no data is currently available regarding the association of fibrinogen level with the presence and severity of new-onset coronary atherosclerosis assessed by Gensini score (GS), particularly in Han Chinese with a large sample size. METHODS AND RESULTS We studied 2288 consecutive, new-onset subjects undergoing coronary angiography with angina-like chest pain. Clinical and laboratory data were collected. Coronary stenotic lesions were considered to be the incidence of coronary atherosclerosis. The severity of coronary stenosis was determined by the GS system. Data indicated that patients with high GS had significantly elevated fibrinogen level (p<0.001). The prevalence and severity of coronary atherosclerosis were dramatically increased according to fibrinogen tertiles. Spearman correlation analysis revealed a positive association between fibrinogen level and GS (r = 0.138, p<0.001). Multivariate logistic regression analysis demonstrated that plasma fibrinogen level was independently associated with high GS (OR = 1.275, 95% CI 1.082-1.502, p = 0.004) after adjusting for potential confounders. Moreover, fibrinogen level was also independently related to the presence of coronary atherosclerosis (fibrinogen tertile 2: OR = 1.192, 95% CI 0.889-1.598, p = 0.241; tertile 3: OR = 2.003, 95% CI 1.383-2.903, p <0.001) and high GS (fibrinogen tertile 2: OR = 1.079, 95% CI 0.833-1.397, p = 0.565; tertile 3: OR = 1.524, 95% CI 1.155-2.011, p = 0.003) in a dose-dependent manner. Receiver-operating characteristic curve analysis showed that the best fibrinogen cut-off value for predicting the severity of coronary stenosis was 3.21 g/L. CONCLUSIONS Higher fibrinogen level is independently linked with the presence and severity of new-onset coronary atherosclerosis in Han Chinese population.
Collapse
Affiliation(s)
- Yan Zhang
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Cheng-Gang Zhu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Yuan-Lin Guo
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Rui-Xia Xu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Sha Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Qian Dong
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Jian-Jun Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| |
Collapse
|
670
|
Lock-Johansson S, Vestbo J, Sorensen GL. Surfactant protein D, Club cell protein 16, Pulmonary and activation-regulated chemokine, C-reactive protein, and Fibrinogen biomarker variation in chronic obstructive lung disease. Respir Res 2014; 15:147. [PMID: 25425298 PMCID: PMC4256818 DOI: 10.1186/s12931-014-0147-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 11/07/2014] [Indexed: 02/06/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a multifaceted condition that cannot be fully described by the severity of airway obstruction. The limitations of spirometry and clinical history have prompted researchers to investigate a multitude of surrogate biomarkers of disease for the assessment of patients, prediction of risk, and guidance of treatment. The aim of this review is to provide a comprehensive summary of observations for a selection of recently investigated pulmonary inflammatory biomarkers (Surfactant protein D (SP-D), Club cell protein 16 (CC-16), and Pulmonary and activation-regulated chemokine (PARC/CCL-18)) and systemic inflammatory biomarkers (C-reactive protein (CRP) and fibrinogen) with COPD. The relevance of these biomarkers for COPD is discussed in terms of their biological plausibility, their independent association to disease and hard clinical outcomes, their modification by interventions, and whether changes in clinical outcomes are reflected by changes in the biomarker.
Collapse
Affiliation(s)
- Sofie Lock-Johansson
- Institute of Molecular Medicine, University of Southern Denmark, JB Winsloews Vej 25.3, Odense, 5000, Denmark.
| | - Jørgen Vestbo
- Department of Respiratory Medicine, Gentofte Hospital, Hellerup, Denmark.
- Respiratory Research Group, Manchester Academic Science Centre University Hospital South Manchester NHS Foundation Trust Manchester, Manchester, UK.
| | - Grith Lykke Sorensen
- Institute of Molecular Medicine, University of Southern Denmark, JB Winsloews Vej 25.3, Odense, 5000, Denmark.
| |
Collapse
|
671
|
O'Doherty MG, Jørgensen T, Borglykke A, Brenner H, Schöttker B, Wilsgaard T, Siganos G, Kavousi M, Hughes M, Müezzinler A, Holleczek B, Franco OH, Hofman A, Boffetta P, Trichopoulou A, Kee F. Repeated measures of body mass index and C-reactive protein in relation to all-cause mortality and cardiovascular disease: results from the consortium on health and ageing network of cohorts in Europe and the United States (CHANCES). Eur J Epidemiol 2014; 29:887-97. [PMID: 25421782 DOI: 10.1007/s10654-014-9954-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 09/23/2014] [Indexed: 12/11/2022]
Abstract
Obesity has been linked with elevated levels of C-reactive protein (CRP), and both have been associated with increased risk of mortality and cardiovascular disease (CVD). Previous studies have used a single 'baseline' measurement and such analyses cannot account for possible changes in these which may lead to a biased estimation of risk. Using four cohorts from CHANCES which had repeated measures in participants 50 years and older, multivariate time-dependent Cox proportional hazards was used to estimate hazard ratios (HR) and 95 % confidence intervals (CI) to examine the relationship between body mass index (BMI) and CRP with all-cause mortality and CVD. Being overweight (≥25-<30 kg/m(2)) or moderately obese (≥30-<35) tended to be associated with a lower risk of mortality compared to normal (≥18.5-<25): ESTHER, HR (95 % CI) 0.69 (0.58-0.82) and 0.78 (0.63-0.97); Rotterdam, 0.86 (0.79-0.94) and 0.80 (0.72-0.89). A similar relationship was found, but only for overweight in Glostrup, HR (95 % CI) 0.88 (0.76-1.02); and moderately obese in Tromsø, HR (95 % CI) 0.79 (0.62-1.01). Associations were not evident between repeated measures of BMI and CVD. Conversely, increasing CRP concentrations, measured on more than one occasion, were associated with an increasing risk of mortality and CVD. Being overweight or moderately obese is associated with a lower risk of mortality, while CRP, independent of BMI, is positively associated with mortality and CVD risk. If inflammation links CRP and BMI, they may participate in distinct/independent pathways. Accounting for independent changes in risk factors over time may be crucial for unveiling their effects on mortality and disease morbidity.
Collapse
Affiliation(s)
- Mark G O'Doherty
- UKCRC Centre of Excellence for Public Health for Northern Ireland, School of Medicine and Dentistry, Queens University Belfast, Grosvenor Road, Belfast, UK, BT12 6BJ, Northern Ireland,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
672
|
Willeit P, Thompson SG, Agewall S, Bergström G, Bickel H, Catapano AL, Chien KL, de Groot E, Empana JP, Etgen T, Franco OH, Iglseder B, Johnsen SH, Kavousi M, Lind L, Liu J, Mathiesen EB, Norata GD, Olsen MH, Papagianni A, Poppert H, Price JF, Sacco RL, Yanez DN, Zhao D, Schminke U, Bülbül A, Polak JF, Sitzer M, Hofman A, Grigore L, Dörr M, Su TC, Ducimetière P, Xie W, Ronkainen K, Kiechl S, Rundek T, Robertson C, Fagerberg B, Bokemark L, Steinmetz H, Ikram MA, Völzke H, Lin HJ, Plichart M, Tuomainen TP, Desvarieux M, McLachlan S, Schmidt C, Kauhanen J, Willeit J, Lorenz MW, Sander D. Inflammatory markers and extent and progression of early atherosclerosis: Meta-analysis of individual-participant-data from 20 prospective studies of the PROG-IMT collaboration. Eur J Prev Cardiol 2014; 23:194-205. [PMID: 25416041 DOI: 10.1177/2047487314560664] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 10/31/2014] [Indexed: 01/09/2023]
Abstract
BACKGROUND Large-scale epidemiological evidence on the role of inflammation in early atherosclerosis, assessed by carotid ultrasound, is lacking. We aimed to quantify cross-sectional and longitudinal associations of inflammatory markers with common-carotid-artery intima-media thickness (CCA-IMT) in the general population. METHODS Information on high-sensitivity C-reactive protein, fibrinogen, leucocyte count and CCA-IMT was available in 20 prospective cohort studies of the PROG-IMT collaboration involving 49,097 participants free of pre-existing cardiovascular disease. Estimates of associations were calculated within each study and then combined using random-effects meta-analyses. RESULTS Mean baseline CCA-IMT amounted to 0.74 mm (SD = 0.18) and mean CCA-IMT progression over a mean of 3.9 years to 0.011 mm/year (SD = 0.039). Cross-sectional analyses showed positive linear associations between inflammatory markers and baseline CCA-IMT. After adjustment for traditional cardiovascular risk factors, mean differences in baseline CCA-IMT per one-SD higher inflammatory marker were: 0.0082 mm for high-sensitivity C-reactive protein (p < 0.001); 0.0072 mm for fibrinogen (p < 0.001); and 0.0025 mm for leucocyte count (p = 0.033). 'Inflammatory load', defined as the number of elevated inflammatory markers (i.e. in upper two quintiles), showed a positive linear association with baseline CCA-IMT (p < 0.001). Longitudinal associations of baseline inflammatory markers and changes therein with CCA-IMT progression were null or at most weak. Participants with the highest 'inflammatory load' had a greater CCA-IMT progression (p = 0.015). CONCLUSION Inflammation was independently associated with CCA-IMT cross-sectionally. The lack of clear associations with CCA-IMT progression may be explained by imprecision in its assessment within a limited time period. Our findings for 'inflammatory load' suggest important combined effects of the three inflammatory markers on early atherosclerosis.
Collapse
Affiliation(s)
- Peter Willeit
- The Department of Public Health and Primary Care, University of Cambridge, UK Department of Neurology, Medical University Innsbruck, Austria
| | - Simon G Thompson
- The Department of Public Health and Primary Care, University of Cambridge, UK
| | - Stefan Agewall
- Institute of Clinical Sciences, University of Oslo, and the Department of Cardiology, Oslo University Hospital Ullevål, Norway
| | - Göran Bergström
- Wallenberg Laboratory for Cardiovascular Research, University of Gothenburg, Sweden
| | - Horst Bickel
- Department of Psychiatry and Psychotherapy, University Hospital of the Technische Universität München, Germany
| | - Alberico L Catapano
- Department of Pharmacological Sciences, University of Milan, and IRCSS Multimedica Sesto S Giovanni, Milan, Italy
| | - Kuo-Liong Chien
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan Department of Internal Medicine, National Taiwan University, Taipei, Taiwan
| | - Eric de Groot
- Academic Medical Centre, Cardiology and Thoracic Surgery, and Imagelabonline and Cardiovascular, Amsterdam, The Netherlands
| | | | - Thorleif Etgen
- Department of Neurology, Kliniken Südostbayern, Klinikum Traunstein, Germany
| | - Oscar H Franco
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Bernhard Iglseder
- Department of Geriatric Medicine, Paracelsus Medical University, and the Gemeinnützige Salzburger Landeskliniken Betriebsgesellschaft GmbH, Christian-Doppler-Klinik, Salzburg, Austria
| | - Stein H Johnsen
- Department of Neurology and Neurophysiology, University Hospital of Northern Norway, and the Department of Clinical Medicine, University of Tromsø, Tromsø, Norway
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Lars Lind
- Department of Medicine, Uppsala University, Sweden
| | - Jing Liu
- Department of Epidemiology, Beijing Institute of Heart, Lung and Blood Vessel Diseases, PR China
| | - Ellisiv B Mathiesen
- Department of Neurology and Neurophysiology, University Hospital of Northern Norway, and the Department of Clinical Medicine, University of Tromsø, Tromsø, Norway
| | - Giuseppe D Norata
- Department of Pharmacological Sciences, University of Milan, and the SISA Centre for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy
| | - Michael H Olsen
- Department of Endocrinology, Centre for Individualized Medicine in Arterial Diseases, Odense University Hospital, Denmark
| | - Aikaterini Papagianni
- Department of Nephrology, Aristotle University of Thessaloniki, Hippokration General Hospital, Greece
| | - Holger Poppert
- Department of Neurology, University Hospital of the Technische Universität München, Germany
| | - Jackie F Price
- Centre for Population Health Sciences, University of Edinburgh, UK
| | - Ralph L Sacco
- Department of Neurology, Miller School of Medicine, University of Miami, FL, USA
| | - David N Yanez
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Dong Zhao
- Department of Epidemiology, Beijing Institute of Heart, Lung and Blood Vessel Diseases, PR China
| | - Ulf Schminke
- Department of Neurology, Greifswald University Clinic, Germany
| | - Alpaslan Bülbül
- Department of Neurology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Joseph F Polak
- Tufts University School of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Matthias Sitzer
- Department of Neurology, University Hospital Frankfurt, Frankfurt am Main, Germany Department of Neurology, Klinikum Herford, Germany
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| | - Liliana Grigore
- Department of Pharmacological Sciences, University of Milan, and the SISA Centre for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy
| | - Marcus Dörr
- Department B for Internal Medicine, University Medicine Greifswald, and the German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Germany
| | - Ta-Chen Su
- Department of Internal Medicine, National Taiwan University, Taipei, Taiwan
| | | | - Wuxiang Xie
- Department of Epidemiology, Beijing Institute of Heart, Lung and Blood Vessel Diseases, PR China
| | - Kimmo Ronkainen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Stefan Kiechl
- Department of Neurology, Medical University Innsbruck, Austria
| | - Tatjana Rundek
- Department of Neurology, Miller School of Medicine, University of Miami, FL, USA
| | | | - Björn Fagerberg
- Wallenberg Laboratory for Cardiovascular Research, University of Gothenburg, Sweden
| | - Lena Bokemark
- Wallenberg Laboratory for Cardiovascular Research, University of Gothenburg, Sweden
| | - Helmuth Steinmetz
- Department of Neurology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Henry Völzke
- Institute for Community Medicine, SHIP/Clinical-Epidemiological Research, Greifswald, Germany
| | - Hung-Ju Lin
- Department of Internal Medicine, National Taiwan University, Taipei, Taiwan Health Management Centre, National Taiwan University Hospital, Taipei, Taiwan
| | - Matthieu Plichart
- INSERM, U970, Université Paris Descartes, France Gerontology Department, Broca Hospital, Paris, France
| | - Tomi-Pekka Tuomainen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Moise Desvarieux
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, USA, and the École des Hautes Études en Santé Publique, and INSERM U738, Paris, France
| | - Stela McLachlan
- Centre for Population Health Sciences, University of Edinburgh, UK
| | - Caroline Schmidt
- Wallenberg Laboratory for Cardiovascular Research, University of Gothenburg, Sweden
| | - Jussi Kauhanen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Johann Willeit
- Department of Neurology, Medical University Innsbruck, Austria
| | - Matthias W Lorenz
- Department of Neurology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Dirk Sander
- Department of Neurology, Benedictus Krankenhaus Tutzing and Feldafing, Tutzing, Germany and Technische Universität München, Germany
| | | |
Collapse
|
673
|
Desai CS, Blumenthal RS, Greenland P. Screening low-risk individuals for coronary artery disease. Curr Atheroscler Rep 2014; 16:402. [PMID: 24522859 DOI: 10.1007/s11883-014-0402-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A large proportion of cardiovascular events occur in individuals classified by traditional risk factors as "low-risk." Efforts to improve early detection of coronary artery disease among low-risk individuals, or to improve risk assessment, might be justified by this large population burden. The most promising tests for improving risk assessment, or early detection, include the coronary artery calcium (CAC) score, the ankle-brachial index (ABI), and the high-sensitivity C-reactive protein (hsCRP). Data regarding the role of additional testing in low-risk populations to improve early detection or to enhance risk assessment are sparse but suggest that CAC and ABI may be helpful for improving risk classification and detecting the higher-risk people from among those at lower risk. However, in the absence of clinical trials in this patient population, such as has recently been proposed, we do not recommend routine use of any additional testing or screening in low-risk individuals at this time.
Collapse
Affiliation(s)
- Chintan S Desai
- Johns Hopkins Hospital, Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA,
| | | | | |
Collapse
|
674
|
Reverri EJ, Morrissey BM, Cross CE, Steinberg FM. Inflammation, oxidative stress, and cardiovascular disease risk factors in adults with cystic fibrosis. Free Radic Biol Med 2014; 76:261-77. [PMID: 25172163 DOI: 10.1016/j.freeradbiomed.2014.08.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 07/31/2014] [Accepted: 08/05/2014] [Indexed: 12/21/2022]
Abstract
Cystic fibrosis (CF) represents one of a number of localized lung and non-lung diseases with an intense chronic inflammatory component associated with evidence of systemic oxidative stress. Many of these chronic inflammatory diseases are accompanied by an array of atherosclerotic processes and cardiovascular disease (CVD), another condition strongly related to inflammation and oxidative stress. As a consequence of a dramatic increase in long-lived patients with CF in recent decades, the specter of CVD must be considered in these patients who are now reaching middle age and beyond. Buttressed by recent data documenting that CF patients exhibit evidence of endothelial dysfunction, a recognized precursor of atherosclerosis and CVD, the spectrum of risk factors for CVD in CF is reviewed here. Epidemiological data further characterizing the presence and extent of atherogenic processes in CF patients would seem important to obtain. Such studies should further inform and offer mechanistic insights into how other chronic inflammatory diseases potentiate the processes leading to CVDs.
Collapse
Affiliation(s)
- Elizabeth J Reverri
- Department of Nutrition, University of California Davis, One Shields Avenue, 3135 Meyer Hall, Davis, CA 95616, USA
| | - Brian M Morrissey
- Adult Cystic Fibrosis Clinic and Division of Pulmonary-Critical Care Medicine, University of California Davis Medical Center, 4150 V Street, Sacramento, CA 95817, USA
| | - Carroll E Cross
- Adult Cystic Fibrosis Clinic and Division of Pulmonary-Critical Care Medicine, University of California Davis Medical Center, 4150 V Street, Sacramento, CA 95817, USA.
| | - Francene M Steinberg
- Department of Nutrition, University of California Davis, One Shields Avenue, 3135 Meyer Hall, Davis, CA 95616, USA
| |
Collapse
|
675
|
Soedamah-Muthu SS, Vergouwe Y, Costacou T, Miller RG, Zgibor J, Chaturvedi N, Snell-Bergeon JK, Maahs DM, Rewers M, Forsblom C, Harjutsalo V, Groop PH, Fuller JH, Moons KGM, Orchard TJ. Predicting major outcomes in type 1 diabetes: a model development and validation study. Diabetologia 2014; 57:2304-14. [PMID: 25186291 PMCID: PMC4399797 DOI: 10.1007/s00125-014-3358-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 07/17/2014] [Indexed: 10/24/2022]
Abstract
AIMS/HYPOTHESIS Type 1 diabetes is associated with a higher risk of major vascular complications and death. A reliable method that predicted these outcomes early in the disease process would help in risk classification. We therefore developed such a prognostic model and quantified its performance in independent cohorts. METHODS Data were analysed from 1,973 participants with type 1 diabetes followed for 7 years in the EURODIAB Prospective Complications Study. Strong prognostic factors for major outcomes were combined in a Weibull regression model. The performance of the model was tested in three different prospective cohorts: the Pittsburgh Epidemiology of Diabetes Complications study (EDC, n = 554), the Finnish Diabetic Nephropathy study (FinnDiane, n = 2,999) and the Coronary Artery Calcification in Type 1 Diabetes study (CACTI, n = 580). Major outcomes included major CHD, stroke, end-stage renal failure, amputations, blindness and all-cause death. RESULTS A total of 95 EURODIAB patients with type 1 diabetes developed major outcomes during follow-up. Prognostic factors were age, HbA1c, WHR, albumin/creatinine ratio and HDL-cholesterol level. The discriminative ability of the model was adequate, with a concordance statistic (C-statistic) of 0.74. Discrimination was similar or even better in the independent cohorts, the C-statistics being: EDC, 0.79; FinnDiane, 0.82; and CACTI, 0.73. CONCLUSIONS/INTERPRETATION Our prognostic model, which uses easily accessible clinical features can discriminate between type 1 diabetes patients who have a good or a poor prognosis. Such a prognostic model may be helpful in clinical practice and for risk stratification in clinical trials.
Collapse
Affiliation(s)
- Sabita S Soedamah-Muthu
- Division of Human Nutrition, Wageningen University, Bomenweg 2, PO Box 8129, 6700 EV, Wageningen, the Netherlands,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
676
|
Soluble urokinase plasminogen activator receptor is in contrast to high-sensitive C-reactive-protein associated with coronary artery calcifications in healthy middle-aged subjects. Atherosclerosis 2014; 237:60-6. [DOI: 10.1016/j.atherosclerosis.2014.08.035] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 08/13/2014] [Accepted: 08/15/2014] [Indexed: 12/21/2022]
|
677
|
Savarese G, Rosano GM, Parente A, D'Amore C, Reiner MF, Camici GG, Trimarco B, Perrone-Filardi P. Reduction of C-reactive protein is not associated with reduced cardiovascular risk and mortality in patients treated with statins. A meta-analysis of 22 randomized trials. Int J Cardiol 2014; 177:152-60. [DOI: 10.1016/j.ijcard.2014.09.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 07/30/2014] [Accepted: 09/16/2014] [Indexed: 12/26/2022]
|
678
|
Lafon A, Tala S, Ahossi V, Perrin D, Giroud M, Béjot Y. Association between periodontal disease and non-fatal ischemic stroke: a case-control study. Acta Odontol Scand 2014; 72:687-93. [PMID: 24720864 DOI: 10.3109/00016357.2014.898089] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE This study aimed to investigate the association between clinical and radiological markers of periodontal disease and ischemic stroke and to assess the potential influence of inflammatory response on the observed associations. METHODS A prospective case-control study including a series of 48 cases with a minor ischemic stroke and 47 controls was conducted at the University Hospital of Dijon. Vascular risk factors, clinical dental examination (plaque index, gingival index, percentage of pockets >5 mm, percentage of bleeding on probing (BOP) sites), dental panoramic (bone loss) and biological parameters (CRP, total cholesterol, HDL, LDL, fasting glucose) were collected. Conditional regression analyses were performed to identify factors associated with ischemic stroke. RESULTS The prevalence of hypertension, high CRP and glucose levels and overall odontological variables was higher in stroke patients. In multivariable analyses, hypertension (OR = 12.56; 95% CI = 2.29-69.96, p = 0.004), CRP levels >5 mg/L (OR = 18.54; 95% CI = 2.01-171.17, p = 0.010), BOP (OR = 1.049; 95% CI = 1.012-1.88, p = 0.009) and bone loss >20% (OR = 1.053; 95% CI = 1.017-1.091, p = 0.004) were associated with ischemic stroke. Among stroke patients, there was a non-significant trend towards higher CRP levels in patients with bone loss >20% compared with those with bone loss <20% (8.1 ± 1.27 mg/L vs 3.12 ± 3.14 mg/L, p = 0.25), whereas other biological parameters were very similar between the two groups. CONCLUSION This case-control study demonstrates that periodontal disease, especially markers such as BOP and bone loss, is independently associated with ischemic stroke.
Collapse
Affiliation(s)
- Arnaud Lafon
- University of Champagne-Ardenne , Reims Cedex , France
| | | | | | | | | | | |
Collapse
|
679
|
Casanova G, dos Reis AM, Spritzer PM. Low-dose oral or non-oral hormone therapy: effects on C-reactive protein and atrial natriuretic peptide in menopause. Climacteric 2014; 18:86-93. [PMID: 25017924 DOI: 10.3109/13697137.2014.940309] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To assess the effects of oral low-dose and non-oral hormone therapy (HT) on ultra-sensitive C-reactive protein (CRP), atrial natriuretic peptide (ANP), and cardiovascular risk factors in postmenopause. METHODS In this randomized, cross-over study, 44 recently postmenopausal women, with no clinical evidence of cardiovascular disease, received oral low-dose HT (estradiol 1 mg + drospirenone 2 mg/day) for 3 months. Forty-two patients received non-oral, conventional HT (1.5 mg/day percutaneous 17β-estradiol gel or equivalent for nasal route) for 3 months followed by 200 mg/day micronized progesterone by the vaginal route (14 days during each menstrual period). After 3 months, patients were crossed over without washout. Post-HT vs. pre-HT measures were determined: lipids, glucose, body mass index, waist circumference, fibrinogen, CRP-stratified levels, and ANP levels. The study was registered at clinical trials.gov (NCT01432028). RESULTS The mean age was 51 ± 3 years and the mean time since the menopause was 22 ± 10 months. CRP-stratified high levels decreased in a higher number of non-oral HT patients, who moved to intermediate and low levels (p = 0.02). No effect of HT was observed on ANP levels (baseline 67.4 (18.4-104.5), low-dose oral 43.5 (14.4-95.9), non-oral 39.8 (15.5-67.5) pg/ml). Markers of endothelial function did not worsen with either low-dose oral or non-oral HT: von Willebrand factor (baseline 118 ± 37%, low-dose oral 119 ± 38%, non-oral 108 ± 3%, p < 0.01), fibrinogen (baseline 356 ± 58 mg/dl; low-dose oral 343 ± 77 mg/dl; non-oral 326 ± 71 mg/dl, p < 0.01). CONCLUSIONS Low-dose oral and non-oral HT for 6 months had neutral or beneficial effects in recently postmenopausal women with no clinical evidence of cardiovascular disease.
Collapse
Affiliation(s)
- G Casanova
- * Gynecological Endocrinology Unit, Division of Endocrinology, Hospital de Clínicas de Porto Alegre , Porto Alegre , Brazil
| | | | | |
Collapse
|
680
|
|
681
|
Cardiovascular Disease Risk Assessment: a Review of Risk Factor-based Algorithms and Assessments of Vascular Health. CURRENT CARDIOVASCULAR RISK REPORTS 2014. [DOI: 10.1007/s12170-014-0419-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
682
|
Takahashi H, Iinuma S, Honma M, Iizuka H. Increased serum C-reactive protein level in Japanese patients of psoriasis with cardio- and cerebrovascular disease. J Dermatol 2014; 41:981-5. [DOI: 10.1111/1346-8138.12632] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 07/15/2014] [Indexed: 12/28/2022]
Affiliation(s)
| | - Shin Iinuma
- Department of Dermatology; Asahikawa Medical University; Asahikawa Japan
| | - Masaru Honma
- Department of Dermatology; Asahikawa Medical University; Asahikawa Japan
| | - Hajime Iizuka
- Department of Dermatology; Asahikawa Medical University; Asahikawa Japan
| |
Collapse
|
683
|
Khandaker GM, Pearson RM, Zammit S, Lewis G, Jones PB. Association of serum interleukin 6 and C-reactive protein in childhood with depression and psychosis in young adult life: a population-based longitudinal study. JAMA Psychiatry 2014; 71:1121-8. [PMID: 25133871 PMCID: PMC4561502 DOI: 10.1001/jamapsychiatry.2014.1332] [Citation(s) in RCA: 597] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
IMPORTANCE Longitudinal studies have linked the systemic inflammatory markers interleukin 6 (IL-6) and C-reactive protein (CRP) with the risk of developing heart disease and diabetes mellitus, which are common comorbidities for depression and psychosis. Recent meta-analyses of cross-sectional studies have reported increased serum levels of these inflammatory markers in depression, first-episode psychosis, and acute psychotic relapse; however, the direction of the association has been unclear. OBJECTIVE To test the hypothesis that higher serum levels of IL-6 and CRP in childhood would increase future risks for depression and psychosis. DESIGN, SETTING, AND PARTICIPANTS The Avon Longitudinal Study of Parents and Children (ALSPAC)is a prospective general population birth cohort study based in Avon County, England. We have studied a subsample of approximately 4500 individuals from the cohort with data on childhood IL-6 and CRP levels and later psychiatric assessments. MEASUREMENT OF EXPOSURE Levels of IL-6 and CRP were measured in nonfasting blood samples obtained in participants at age 9 years. MAIN OUTCOMES AND MEASURES Participants were assessed at age 18 years. Depression was measured using the Clinical Interview Schedule-Revised (CIS-R) and Mood and Feelings Questionnaire (MFQ), thus allowing internal replication; psychotic experiences (PEs) and psychotic disorder were measured by a semistructured interview. RESULTS After adjusting for sex, age, body mass index, ethnicity, social class, past psychological and behavioral problems, and maternal postpartum depression, participants in the top third of IL-6 values compared with the bottom third at age 9 years were more likely to be depressed (CIS-R) at age 18 years (adjusted odds ratio [OR], 1.55; 95% CI, 1.13-2.14). Results using the MFQ were similar. Risks of PEs and of psychotic disorder at age 18 years were also increased with higher IL-6 levels at baseline (adjusted OR, 1.81; 95% CI, 1.01-3.28; and adjusted OR, 2.40; 95% CI, 0.88-6.22, respectively). Higher IL-6 levels in childhood were associated with subsequent risks of depression and PEs in a dose-dependent manner. CONCLUSIONS AND RELEVANCE Higher levels of the systemic inflammatory marker IL-6 in childhood are associated with an increased risk of developing depression and psychosis in young adulthood. Inflammatory pathways may provide important new intervention and prevention targets for these disorders. Inflammation might explain the high comorbidity between heart disease, diabetes mellitus, depression, and schizophrenia.
Collapse
Affiliation(s)
- Golam M Khandaker
- Department of Psychiatry, University of Cambridge, Cambridge, England2National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, England3Collaboration for Leadership in Applied Health Research and Care, East of England, Cambri
| | - Rebecca M Pearson
- Centre for Mental Health, Addiction and Suicide Research, School of Social and Community Medicine, University of Bristol, Bristol, England
| | - Stanley Zammit
- Centre for Mental Health, Addiction and Suicide Research, School of Social and Community Medicine, University of Bristol, Bristol, England6Institute of Psychological Medicine and Clinical Neurosciences, Medical Research Council Centre for Neuropsychiatric
| | - Glyn Lewis
- Centre for Mental Health, Addiction and Suicide Research, School of Social and Community Medicine, University of Bristol, Bristol, England7Division of Psychiatry, University College London, London, England
| | - Peter B Jones
- Department of Psychiatry, University of Cambridge, Cambridge, England2National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, England3Collaboration for Leadership in Applied Health Research and Care, East of England, Cambri
| |
Collapse
|
684
|
Cheng A, Zhang Y, Blasco-Colmenares E, Dalal D, Butcher B, Norgard S, Eldadah Z, Ellenbogen KA, Dickfeld T, Spragg DD, Marine JE, Guallar E, Tomaselli GF. Protein biomarkers identify patients unlikely to benefit from primary prevention implantable cardioverter defibrillators: findings from the Prospective Observational Study of Implantable Cardioverter Defibrillators (PROSE-ICD). Circ Arrhythm Electrophysiol 2014; 7:1084-91. [PMID: 25273351 DOI: 10.1161/circep.113.001705] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Primary prevention implantable cardioverter defibrillators (ICDs) reduce all-cause mortality, but the benefits are heterogeneous. Current risk stratification based on left ventricular ejection fraction has limited discrimination power. We hypothesize that biomarkers for inflammation, neurohumoral activation, and cardiac injury can predict appropriate shocks and all-cause mortality in patients with primary prevention ICDs. METHODS AND RESULTS The Prospective Observational Study of Implantable Cardioverter Defibrillators (PROSe-ICD) enrolled 1189 patients with systolic heart failure who underwent ICD implantation for primary prevention of sudden cardiac death. The primary end point was an ICD shock for adjudicated ventricular tachyarrhythmia. The secondary end point was all-cause mortality. After a median follow-up of 4.0 years, 137 subjects experienced an appropriate ICD shock and 343 participants died (incidence rates of 3.2 and 5.8 per 100 person-years, respectively). In multivariable-adjusted models, higher interleukin-6 levels increased the risk of appropriate ICD shocks. In contrast, C-reactive protein, interleukin-6, tumor necrosis factor-α receptor II, pro-brain natriuretic peptide (pro-BNP), and cardiac troponin T showed significant linear trends for increased risk of all-cause mortality across quartiles. A score combining these 5 biomarkers identified patients who were much more likely to die than to receive an appropriate shock from the ICD. CONCLUSIONS An increase in serum biomarkers of inflammation, neurohumoral activation, and myocardial injury increased the risk for death but poorly predicted the likelihood of an ICD shock. These findings highlight the potential importance of serum-based biomarkers in identifying patients who are unlikely to benefit from primary prevention ICDs. CLINICAL TRIAL REGISTRATION URL clinicaltrials.gov; Unique Identifier: NCT00733590.
Collapse
Affiliation(s)
- Alan Cheng
- From the Department of Medicine (A.C., E.B.-C., D.D., B.B., S.N., D.D.S., J.E.M., G.F.T.) and the Welch Center for Prevention, Epidemiology and Clinical Research (Y.Z., E.G.), Johns Hopkins Medical Institutions, Baltimore, MD; Washington Hospital Center, DC (Z.E.); Medical College of Virginia, Richmond (K.A.E.); and University of Maryland, Baltimore (T.D.)
| | - Yiyi Zhang
- From the Department of Medicine (A.C., E.B.-C., D.D., B.B., S.N., D.D.S., J.E.M., G.F.T.) and the Welch Center for Prevention, Epidemiology and Clinical Research (Y.Z., E.G.), Johns Hopkins Medical Institutions, Baltimore, MD; Washington Hospital Center, DC (Z.E.); Medical College of Virginia, Richmond (K.A.E.); and University of Maryland, Baltimore (T.D.)
| | - Elena Blasco-Colmenares
- From the Department of Medicine (A.C., E.B.-C., D.D., B.B., S.N., D.D.S., J.E.M., G.F.T.) and the Welch Center for Prevention, Epidemiology and Clinical Research (Y.Z., E.G.), Johns Hopkins Medical Institutions, Baltimore, MD; Washington Hospital Center, DC (Z.E.); Medical College of Virginia, Richmond (K.A.E.); and University of Maryland, Baltimore (T.D.)
| | - Darshan Dalal
- From the Department of Medicine (A.C., E.B.-C., D.D., B.B., S.N., D.D.S., J.E.M., G.F.T.) and the Welch Center for Prevention, Epidemiology and Clinical Research (Y.Z., E.G.), Johns Hopkins Medical Institutions, Baltimore, MD; Washington Hospital Center, DC (Z.E.); Medical College of Virginia, Richmond (K.A.E.); and University of Maryland, Baltimore (T.D.)
| | - Barbara Butcher
- From the Department of Medicine (A.C., E.B.-C., D.D., B.B., S.N., D.D.S., J.E.M., G.F.T.) and the Welch Center for Prevention, Epidemiology and Clinical Research (Y.Z., E.G.), Johns Hopkins Medical Institutions, Baltimore, MD; Washington Hospital Center, DC (Z.E.); Medical College of Virginia, Richmond (K.A.E.); and University of Maryland, Baltimore (T.D.)
| | - Sanaz Norgard
- From the Department of Medicine (A.C., E.B.-C., D.D., B.B., S.N., D.D.S., J.E.M., G.F.T.) and the Welch Center for Prevention, Epidemiology and Clinical Research (Y.Z., E.G.), Johns Hopkins Medical Institutions, Baltimore, MD; Washington Hospital Center, DC (Z.E.); Medical College of Virginia, Richmond (K.A.E.); and University of Maryland, Baltimore (T.D.)
| | - Zayd Eldadah
- From the Department of Medicine (A.C., E.B.-C., D.D., B.B., S.N., D.D.S., J.E.M., G.F.T.) and the Welch Center for Prevention, Epidemiology and Clinical Research (Y.Z., E.G.), Johns Hopkins Medical Institutions, Baltimore, MD; Washington Hospital Center, DC (Z.E.); Medical College of Virginia, Richmond (K.A.E.); and University of Maryland, Baltimore (T.D.)
| | - Kenneth A Ellenbogen
- From the Department of Medicine (A.C., E.B.-C., D.D., B.B., S.N., D.D.S., J.E.M., G.F.T.) and the Welch Center for Prevention, Epidemiology and Clinical Research (Y.Z., E.G.), Johns Hopkins Medical Institutions, Baltimore, MD; Washington Hospital Center, DC (Z.E.); Medical College of Virginia, Richmond (K.A.E.); and University of Maryland, Baltimore (T.D.)
| | - Timm Dickfeld
- From the Department of Medicine (A.C., E.B.-C., D.D., B.B., S.N., D.D.S., J.E.M., G.F.T.) and the Welch Center for Prevention, Epidemiology and Clinical Research (Y.Z., E.G.), Johns Hopkins Medical Institutions, Baltimore, MD; Washington Hospital Center, DC (Z.E.); Medical College of Virginia, Richmond (K.A.E.); and University of Maryland, Baltimore (T.D.)
| | - David D Spragg
- From the Department of Medicine (A.C., E.B.-C., D.D., B.B., S.N., D.D.S., J.E.M., G.F.T.) and the Welch Center for Prevention, Epidemiology and Clinical Research (Y.Z., E.G.), Johns Hopkins Medical Institutions, Baltimore, MD; Washington Hospital Center, DC (Z.E.); Medical College of Virginia, Richmond (K.A.E.); and University of Maryland, Baltimore (T.D.)
| | - Joseph E Marine
- From the Department of Medicine (A.C., E.B.-C., D.D., B.B., S.N., D.D.S., J.E.M., G.F.T.) and the Welch Center for Prevention, Epidemiology and Clinical Research (Y.Z., E.G.), Johns Hopkins Medical Institutions, Baltimore, MD; Washington Hospital Center, DC (Z.E.); Medical College of Virginia, Richmond (K.A.E.); and University of Maryland, Baltimore (T.D.)
| | - Eliseo Guallar
- From the Department of Medicine (A.C., E.B.-C., D.D., B.B., S.N., D.D.S., J.E.M., G.F.T.) and the Welch Center for Prevention, Epidemiology and Clinical Research (Y.Z., E.G.), Johns Hopkins Medical Institutions, Baltimore, MD; Washington Hospital Center, DC (Z.E.); Medical College of Virginia, Richmond (K.A.E.); and University of Maryland, Baltimore (T.D.)
| | - Gordon F Tomaselli
- From the Department of Medicine (A.C., E.B.-C., D.D., B.B., S.N., D.D.S., J.E.M., G.F.T.) and the Welch Center for Prevention, Epidemiology and Clinical Research (Y.Z., E.G.), Johns Hopkins Medical Institutions, Baltimore, MD; Washington Hospital Center, DC (Z.E.); Medical College of Virginia, Richmond (K.A.E.); and University of Maryland, Baltimore (T.D.).
| |
Collapse
|
685
|
Mayers JR, Wu C, Clish CB, Kraft P, Torrence ME, Fiske BP, Yuan C, Bao Y, Townsend MK, Tworoger SS, Davidson SM, Papagiannakopoulos T, Yang A, Dayton TL, Ogino S, Stampfer MJ, Giovannucci EL, Qian ZR, Rubinson DA, Ma J, Sesso HD, Gaziano JM, Cochrane BB, Liu S, Wactawski–Wende J, Manson JE, Pollak MN, Kimmelman AC, Souza A, Pierce K, Wang TJ, Gerszten RE, Fuchs CS, Heiden MGV, Wolpin BM. Elevation of circulating branched-chain amino acids is an early event in human pancreatic adenocarcinoma development. Nat Med 2014; 20:1193-1198. [PMID: 25261994 PMCID: PMC4191991 DOI: 10.1038/nm.3686] [Citation(s) in RCA: 493] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 08/05/2014] [Indexed: 12/12/2022]
Abstract
Most patients with pancreatic ductal adenocarcinoma (PDAC) are diagnosed with advanced disease and survive less than 12 months. PDAC has been linked with obesity and glucose intolerance, but whether changes in circulating metabolites are associated with early cancer progression is unknown. To better understand metabolic derangements associated with early disease, we profiled metabolites in prediagnostic plasma from individuals with pancreatic cancer (cases) and matched controls from four prospective cohort studies. We find that elevated plasma levels of branched-chain amino acids (BCAAs) are associated with a greater than twofold increased risk of future pancreatic cancer diagnosis. This elevated risk was independent of known predisposing factors, with the strongest association observed among subjects with samples collected 2 to 5 years before diagnosis, when occult disease is probably present. We show that plasma BCAAs are also elevated in mice with early-stage pancreatic cancers driven by mutant Kras expression but not in mice with Kras-driven tumors in other tissues, and that breakdown of tissue protein accounts for the increase in plasma BCAAs that accompanies early-stage disease. Together, these findings suggest that increased whole-body protein breakdown is an early event in development of PDAC.
Collapse
Affiliation(s)
- Jared R. Mayers
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA
| | - Chen Wu
- Department of Etiology and Carcinogenesis, Cancer Institute and Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Department of Medical Oncology, Dana–Farber Cancer Institute and Harvard Medical School, Boston, MA
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
| | - Clary B. Clish
- Broad Institute of MIT and Harvard University, Cambridge, MA
| | - Peter Kraft
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
- Department of Biostatistics, Harvard School of Public Health, Boston, MA
| | - Margaret E. Torrence
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA
| | - Brian P. Fiske
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA
| | - Chen Yuan
- Department of Medical Oncology, Dana–Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Ying Bao
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Mary K. Townsend
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Shelley S. Tworoger
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Shawn M. Davidson
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA
| | - Thales Papagiannakopoulos
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA
| | - Annan Yang
- Division of Genomic Stability and DNA repair, Department of Radiation Oncology, Dana– Farber Cancer Institute, Boston, MA 02215
| | - Talya L. Dayton
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA
| | - Shuji Ogino
- Department of Medical Oncology, Dana–Farber Cancer Institute and Harvard Medical School, Boston, MA
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Meir J. Stampfer
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
- Department of Nutrition, Harvard School of Public Health, Boston, MA
| | - Edward L. Giovannucci
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
- Department of Nutrition, Harvard School of Public Health, Boston, MA
| | - Zhi Rong Qian
- Department of Medical Oncology, Dana–Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Douglas A. Rubinson
- Department of Medical Oncology, Dana–Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Jing Ma
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Howard D. Sesso
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA
| | - John Michael Gaziano
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System
| | | | - Simin Liu
- Departments of Epidemiology and Medicine, Brown University, Providence, RI
| | - Jean Wactawski–Wende
- Department of Social and Preventive Medicine, University at Buffalo, SUNY, Buffalo, NY
| | - JoAnn E. Manson
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA
| | - Michael N. Pollak
- Departments of Oncology and Medicine, McGill University, Montreal, QC, Canada
| | - Alec C. Kimmelman
- Division of Genomic Stability and DNA repair, Department of Radiation Oncology, Dana– Farber Cancer Institute, Boston, MA 02215
| | - Amanda Souza
- Broad Institute of MIT and Harvard University, Cambridge, MA
| | - Kerry Pierce
- Broad Institute of MIT and Harvard University, Cambridge, MA
| | - Thomas J. Wang
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN
| | - Robert E. Gerszten
- Broad Institute of MIT and Harvard University, Cambridge, MA
- Cardiology Division, Massachusetts General Hospital, and Harvard Medical School, Boston, MA
| | - Charles S. Fuchs
- Department of Medical Oncology, Dana–Farber Cancer Institute and Harvard Medical School, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Matthew G. Vander Heiden
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA
- Department of Medical Oncology, Dana–Farber Cancer Institute and Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard University, Cambridge, MA
| | - Brian M. Wolpin
- Department of Medical Oncology, Dana–Farber Cancer Institute and Harvard Medical School, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA
| |
Collapse
|
686
|
Segal HC, Burgess AI, Poole DL, Mehta Z, Silver LE, Rothwell PM. Population-based study of blood biomarkers in prediction of subacute recurrent stroke. Stroke 2014; 45:2912-7. [PMID: 25158774 PMCID: PMC5380212 DOI: 10.1161/strokeaha.114.005592] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 07/24/2014] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND PURPOSE Risk of recurrent stroke is high in the first few weeks after transient ischemic attack or stroke and clinical risk prediction tools have only limited accuracy, particularly after the hyperacute phase. Previous studies of the predictive value of biomarkers have been small, been done in selected populations, and have not concentrated on the acute phase or on intensively treated populations. We aimed to determine the predictive value of a panel of blood biomarkers in intensively treated patients early after transient ischemic attack and stroke. METHODS We studied 14 blood biomarkers related to inflammation, thrombosis, atherogenesis, and cardiac or neuronal cell damage in early transient ischemic attack or ischemic stroke in a population-based study (Oxford Vascular Study). Biomarker levels were related to 90-day risk of recurrent stroke as hazard ratio (95% confidence interval) per decile increase, adjusted for age and sex. RESULTS Among 1292 eligible patients, there were 53 recurrent ischemic strokes within 90 days. There were moderate correlations (r=0.40-0.61; P<0.0001) between the inflammatory biomarkers and between the cell damage and thrombotic subsets. Associations with risk of early recurrent stroke were weak, with significant associations limited to interleukin-6 (adjusted hazard ratio, 1.12; 1.01-1.24; P=0.033) and C-reactive protein (adjusted hazard ratio, 1.15; 1.02-1.30; P=0.022) after adjusting for age, sex, hypertension, smoking, and diabetes mellitus although P-selectin seemed to predict stroke after transient ischemic attack (adjusted hazard ratio, 1.28; 1.00-1.63; P=0.046). CONCLUSIONS In the largest study to date, we found limited predictive use for early recurrent stroke for a panel of inflammatory, thrombotic, and cell damage biomarkers.
Collapse
Affiliation(s)
- Helen C Segal
- From the Stroke Prevention Research Unit, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom
| | - Annette I Burgess
- From the Stroke Prevention Research Unit, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom
| | - Debbie L Poole
- From the Stroke Prevention Research Unit, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom
| | - Ziyah Mehta
- From the Stroke Prevention Research Unit, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom
| | - Louise E Silver
- From the Stroke Prevention Research Unit, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom
| | - Peter M Rothwell
- From the Stroke Prevention Research Unit, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom.
| |
Collapse
|
687
|
Case SM, Stewart JC. Race/ethnicity moderates the relationship between depressive symptom severity and C-reactive protein: 2005-2010 NHANES data. Brain Behav Immun 2014; 41:101-8. [PMID: 24859042 DOI: 10.1016/j.bbi.2014.04.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 02/28/2014] [Accepted: 04/10/2014] [Indexed: 12/15/2022] Open
Abstract
Because few studies have examined depression facets or potential moderators of the depression-inflammation relationship, our aims were to determine whether particular depressive symptom clusters are more strongly associated with C-reactive protein (CRP) levels and whether race/ethnicity moderates these relationships. We examined data from 10,149 adults representative of the U.S. population (4858 non-Hispanic White, 1978 non-Hispanic Black, 2260 Mexican American, 1053 Other Hispanic) who participated in the cross-sectional National Health and Nutrition Examination Survey between 2005 and 2010. Depressive symptoms were assessed by the Patient Health Questionnaire-9, and high-sensitivity serum CRP was quantified by latex-enhanced nephelometry. Total (p<.001), somatic (p<.001), and nonsomatic (p=.001) depressive symptoms were each positively related to serum CRP in individual models. However, in the simultaneous model that included both symptom clusters, somatic symptoms (p<.001), but not nonsomatic symptoms (p=.98), remained associated with serum CRP. Evidence of moderation by race/ethnicity was also observed, as six of the nine depressive symptoms×race/ethnicity interactions were significant (ps<.05). Among non-Hispanic Whites, the pattern of results was identical to the full sample; only somatic symptoms (p<.001) remained related to serum CRP in the simultaneous model. No relationships between total, somatic, or nonsomatic symptoms and serum CRP were observed among the non-Hispanic Black, Mexican American, or Other Hispanic groups. Our findings indicate that the link between depressive symptoms and systemic inflammation may be due to the somatic symptoms of sleep disturbance, fatigue, appetite changes, and psychomotor retardation/agitation and may be strongest among non-Hispanic Whites.
Collapse
Affiliation(s)
- Stephanie M Case
- School of Psychological Sciences, University of Indianapolis, 1400 East Hanna Avenue, Indianapolis, IN 46227, United States.
| | - Jesse C Stewart
- Department of Psychology, Indiana University-Purdue University Indianapolis, 402 N. Blackford St., LD 100E, Indianapolis, IN 46202, United States.
| |
Collapse
|
688
|
Sharma M, Ganguly N. Burden of cardiovascular diseases in Indians: Estimating trends of coronary artery disease and using low cost risk screening tools. APOLLO MEDICINE 2014. [DOI: 10.1016/j.apme.2014.07.002] [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] Open
|
689
|
Zhang Y, Zhu CG, Xu RX, Li S, Guo YL, Sun J, Li JJ. Relation of circulating PCSK9 concentration to fibrinogen in patients with stable coronary artery disease. J Clin Lipidol 2014; 8:494-500. [PMID: 25234562 DOI: 10.1016/j.jacl.2014.07.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 07/02/2014] [Accepted: 07/02/2014] [Indexed: 11/22/2022]
Abstract
BACKGROUND Both proprotein convertase subtilisin/kexin type 9 (PCSK9) and fibrinogen have been established as novel markers for atherosclerotic diseases. However, no data are available regarding the relationship between circulating PCSK9 and fibrinogen concentration up to now. OBJECTIVE To explore the potential link of the circulating PCSK9 concentration to fibrinogen in patients with stable coronary artery disease (CAD). METHODS We studied 219 eligible consecutive patients with angiographically proven stable CAD. Baseline clinical and laboratory data were collected. Plasma PCSK9 concentration was measured by enzyme-linked immunosorbent assay. High-sensitivity C-reactive protein (hs-CRP), erythrocyte sedimentation rate, D-dimer, and albumin were also measured in all subjects as inflammatory markers. The relation of the circulating PCSK9 concentration to fibrinogen was evaluated. RESULTS The data indicated that the patients with high PCSK9 concentration tended to have higher fibrinogen levels according to PCSK9 tertiles (P = .037). Spearman correlation analysis revealed a positive relation between plasma PCSK9 concentration and fibrinogen (r = 0.211, P = .002). Additionally, the circulating PCSK9 concentration also correlated positively with total cholesterol, low-density lipoprotein cholesterol, and hs-CRP levels (r = 0.333, P < .001; r = 0.302, P < .001; r = 0.153, P = .023, respectively). In the stepwise multivariate regression analysis, the association between PCSK9 and fibrinogen remained significant (β = 0.168, P = .011) after adjustment for conventional cardiovascular risk factors including lipid profiles, hs-CRP, and D-dimer. CONCLUSION The present study first demonstrated that the circulating PCSK9 concentration was positively associated with fibrinogen in patients with stable CAD, suggesting that the interactions of PCSK9 and fibrinogen in the status of atherosclerosis may need further investigation.
Collapse
Affiliation(s)
- Yan Zhang
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Cheng-Gang Zhu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Rui-Xia Xu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Sha Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yuan-Lin Guo
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jing Sun
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jian-Jun Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.
| |
Collapse
|
690
|
Ndrepepa G, Braun S, Tada T, King L, Cassese S, Fusaro M, Keta D, Kastrati A, Schmidt R. Comparative prognostic value of C-reactive protein & fibrinogen in patients with coronary artery disease. Indian J Med Res 2014; 140:392-400. [PMID: 25366207 PMCID: PMC4248386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND & OBJECTIVES The comparative prognostic value of C-reactive protein (CRP) and fibrinogen for cardiovascular events has been inconclusively investigated. t0 his study was carried out to compare the prognostic value of CRP versus fibrinogen in patients with coronary artery disease (CAD). METHODS The study included 13,100 patients with coronary angiography-confirmed CAD. Plasma CRP and fibrinogen levels were measured before angiography in all patients. The levels of CRP>3 mg/l and fibrinogen>350 mg/dl were considered as elevated. The primary outcome was 1-year all-cause mortality. RESULTS Patients were divided into four groups: patients with CRP≤3 mg/l and fibrinogen ≤350 mg/dl (n=4206); patients with CRP≤3 mg/l and fibrinogen >350 mg/dl (n=3132); patients with CRP>3 mg/l and fibrinogen ≤ 350 mg/dl (n=1273) and CRP >3 mg/l and patients with fibrinogen >350 mg/dl (n=4489). There were 634 deaths: 75 deaths in patients with CRP ≤3 mg/l and fibrinogen ≤350 mg/dl, 91 deaths in patients with CRP ≤3 mg/l and fibrinogen >350 mg/dl, 87 deaths in patients with CRP >3 mg/l and fibrinogen ≤350 mg/dl and 381 deaths in patients with CRP >3 mg/l and fibrinogen >350 mg/dl (Kaplan-Meier estimates of all-cause mortality, 1.8, 3.0, 7.0 and 8.7 %, log-rank test P<0.001). The multivariate analysis showed that CRP [adjusted hazard ratio (HR)=1.31, 95% confidence interval (CI) 1.18-1.45, for each standard deviation increase in the logarithmic scale] but not fibrinogen [adjusted HR=0.99 (0.90-1.09), for each standard deviation increase in the logarithmic scale] was an independent correlate of mortality. INTERPRETATION & CONCLUSIONS The findings indicated that in patients with CAD, CRP was a better predictor of mortality than fibrinogen and offered prognostic information beyond that provided by the conventional cardiovascular risk factors.
Collapse
Affiliation(s)
- Gjin Ndrepepa
- German Heart Centre, Technical University, Munich, Germany,Reprint requests: Dr Gjin Ndrepepa, Deutsches Herzzentrum, Lazarettstrasse 36, 80636 München, Germany e-mail:
| | - Siegmund Braun
- German Heart Centre, Technical University, Munich, Germany
| | - Tomohisa Tada
- German Heart Centre, Technical University, Munich, Germany
| | - Lamin King
- German Heart Centre, Technical University, Munich, Germany
| | | | | | - Dritan Keta
- Medical Clinic I, Landshut-Achdorf Hospital, Landshut, Germany
| | - Adnan Kastrati
- German Heart Centre, Technical University, Munich, Germany
| | | |
Collapse
|
691
|
Lee Y, Kang D, Lee SA. Effect of dietary patterns on serum C-reactive protein level. Nutr Metab Cardiovasc Dis 2014; 24:1004-1011. [PMID: 24998076 DOI: 10.1016/j.numecd.2014.05.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 04/02/2014] [Accepted: 05/07/2014] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIMS C-reactive protein (CRP) is a marker of inflammation which has been shown in multiple prospective epidemiological studies to predict the risk of cardiovascular disease and metabolic syndrome. Dietary patterns may influence the risk of diseases through the effects of CRP on inflammation. The aim of this study was to examine the relationship of dietary patterns with CRP in blood, taking into consideration gender and blood pressure. METHODS AND RESULTS The present cross-sectional analysis included 7574 participants from a large, population-based study of middle-aged Koreans. Four dietary patterns, including 'fruit', 'vegetable', 'meat' and 'coffee', were derived using factor analysis. Multiple linear regressions were used to evaluate the association between dietary patterns and CRP after adjusting for age, sex, education, waist circumference, smoke status and alcohol consumption. We found that the 'vegetable' pattern was inversely associated with CRP (Ptrend = 0.01). The adjusted mean CRP was 0.04 mg/dl lower (95% CI -0.07, -0.01) for subjects in the highest quartile of the 'vegetable' pattern compared with those in the lowest quartile. This inverse association was more pronounced in men with hypertensive blood; CRP concentrations across increasing quartile categories of the 'vegetable' pattern score were 0.1, 0.14, and 0.15 mg/dl reduction compared to the 1st quartile (the lowest quartile) (Ptrend = 0.04, Plinearity = 0.02). CONCLUSIONS The inverse association of the 'vegetable' pattern with CRP was assessed, and the association appeared to be more predominant in men with hypertensive blood pressure.
Collapse
Affiliation(s)
- Y Lee
- Department of Biomedical Sciences, Seoul National University, Seoul, Republic of Korea; Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - D Kang
- Department of Biomedical Sciences, Seoul National University, Seoul, Republic of Korea; Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - S-A Lee
- Department of Preventive Medicine, Kangwon National University School of Medicine, Gangwon-do, Republic of Korea.
| |
Collapse
|
692
|
Duivenvoorden R, Stroes ESG, Hovingh GK. The value of screening for carotid plaque in patients referred for echocardiography. Can J Cardiol 2014; 30:1148-9. [PMID: 25154805 DOI: 10.1016/j.cjca.2014.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 07/02/2014] [Indexed: 10/25/2022] Open
Affiliation(s)
- Raphael Duivenvoorden
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Erik S G Stroes
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - G Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
693
|
Eckel RH, Cornier MA. Update on the NCEP ATP-III emerging cardiometabolic risk factors. BMC Med 2014; 12:115. [PMID: 25154373 PMCID: PMC4283079 DOI: 10.1186/1741-7015-12-115] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 05/20/2014] [Indexed: 12/31/2022] Open
Abstract
The intent of this review is to update the science of emerging cardiometabolic risk factors that were listed in the National Cholesterol Education Program (NCEP) Adult Treatment Panel-III (ATP-III) report of 2001 (updated in 2004). At the time these guidelines were published, the evidence was felt to be insufficient to recommend these risk factors for routine screening of cardiovascular disease risk. However, the panel felt that prudent use of these biomarkers for patients at intermediate risk of a major cardiovascular event over the subsequent 10 years might help identify patients who needed more aggressive low density lipoprotein (LDL) or non-high density lipoprotein (HDL) cholesterol lowering therapy. While a number of other emerging risk factors have been identified, this review will be limited to assessing the data and recommendations for the use of apolipoprotein B, lipoprotein (a), homocysteine, pro-thrombotic factors, inflammatory factors, impaired glucose metabolism, and measures of subclinical atherosclerotic cardiovascular disease for further cardiovascular disease risk stratification.
Collapse
Affiliation(s)
- Robert H Eckel
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Anschutz Medical Campus, Mail Stop 8106, 12801 E 17th Ave, Aurora, CO 80045 USA
| | - Marc-Andre Cornier
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Anschutz Medical Campus, Mail Stop 8106, 12801 E 17th Ave, Aurora, CO 80045 USA
| |
Collapse
|
694
|
Higueras J, Martín-Ventura JL, Blanco-Colio L, Cristóbal C, Tarín N, Huelmos A, Alonso J, Pello A, Aceña Á, Carda R, Lorenzo Ó, Mahíllo-Fernández I, Asensio D, Almeida P, Rodríguez-Artalejo F, Farré J, López Bescós L, Egido J, Tuñón J. [Impact of plasma pro-B-type natriuretic peptide amino-terminal and galectin-3 levels on the predictive capacity of the LIPID Clinical Risk Scale in stable coronary disease]. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2014; 27:57-63. [PMID: 25127747 DOI: 10.1016/j.arteri.2014.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 06/23/2014] [Indexed: 11/18/2022]
Abstract
INTRODUCTION At present, there is no tool validated by scientific societies for risk stratification of patients with stable coronary artery disease (SCAD). It has been shown that plasma levels of monocyte chemoattractant protein-1 (MCP-1), galectin-3 and pro-B-type natriuretic peptide amino-terminal (NT-proBNP) have prognostic value in this population. OBJECTIVE To analyze the prognostic value of a clinical risk scale published in Long-term Intervention with Pravastatin in Ischemic Disease (LIPID) study and determining its predictive capacity when combined with plasma levels of MCP-1, galectin-3 and NT-proBNP in patients with SCAD. METHODS AND RESULTS A total of 706 patients with SCAD and a history of acute coronary syndrome (ACS) were analyzed over a follow up period of 2.2 ± 0.99 years. The primary endpoint was the occurrence of an ischemic event (any SCA, stroke or transient ischemic attack), heart failure, or death. A clinical risk scale derived from the LIPID study significantly predicted the development of the primary endpoint, with an area under the ROC curve (Receiver Operating Characteristic) of 0.642 (0.579 to 0.705); P<0.001. A composite score was developed by adding the scores of the LIPID and scale decile levels of MCP -1, galectin -3 and NT-proBNP. The predictive value improved with an area under the curve of 0.744 (0.684 to 0.805); P<0.001 (P=0.022 for comparison). A score greater than 21.5 had a sensitivity of 74% and a specificity of 61% for the development of the primary endpoint (P<0.001, log -rank test). CONCLUSION Plasma levels of MCP-1, galectin -3 and NT-proBNP improve the ability of the LIPID clinical scale to predict the prognosis of patients with SCAD.
Collapse
Affiliation(s)
- Javier Higueras
- Servicio de Cardiología, Hospital Clínico Universitario San Carlos, Madrid, España; Universidad Complutense, Madrid, España
| | - José Luis Martín-Ventura
- Universidad Autónoma, Madrid, España; Laboratorio de Patología Vascular, IIS-Fundación Jiménez Díaz, Madrid, España
| | - Luis Blanco-Colio
- Laboratorio de Patología Vascular, IIS-Fundación Jiménez Díaz, Madrid, España
| | - Carmen Cristóbal
- Servicio de Cardiología, Hospital de Fuenlabrada, Fuenlabrada, Madrid, España; Universidad Rey Juan Carlos, Alcorcón, Madrid, España
| | - Nieves Tarín
- Servicio de Cardiología, Hospital Universitario de Móstoles, Móstoles, Madrid, España
| | - Ana Huelmos
- Servicio de Cardiología, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, España
| | - Joaquín Alonso
- Servicio de Cardiología, Hospital de Fuenlabrada, Fuenlabrada, Madrid, España; Universidad Rey Juan Carlos, Alcorcón, Madrid, España
| | - Ana Pello
- Servicio de Cardiología, IIS-Fundación Jiménez Díaz, Madrid, España
| | - Álvaro Aceña
- Servicio de Cardiología, IIS-Fundación Jiménez Díaz, Madrid, España
| | - Rocío Carda
- Servicio de Cardiología, IIS-Fundación Jiménez Díaz, Madrid, España
| | - Óscar Lorenzo
- Universidad Autónoma, Madrid, España; Laboratorio de Patología Vascular, IIS-Fundación Jiménez Díaz, Madrid, España
| | | | - Dolores Asensio
- Laboratorio de bioquímica de la Fundación Jiménez Díaz, Madrid, España
| | - Pedro Almeida
- Servicio de Cardiología, IIS-Fundación Jiménez Díaz, Madrid, España
| | | | - Jerónimo Farré
- Universidad Autónoma, Madrid, España; Servicio de Cardiología, IIS-Fundación Jiménez Díaz, Madrid, España
| | | | - Jesús Egido
- Universidad Autónoma, Madrid, España; Laboratorio de Patología Vascular, IIS-Fundación Jiménez Díaz, Madrid, España; CIBERDEM, Madrid, España
| | - José Tuñón
- Universidad Autónoma, Madrid, España; Laboratorio de Patología Vascular, IIS-Fundación Jiménez Díaz, Madrid, España; Servicio de Cardiología, IIS-Fundación Jiménez Díaz, Madrid, España.
| |
Collapse
|
695
|
DeJarnett N, Conklin DJ, Riggs DW, Myers JA, O'Toole TE, Hamzeh I, Wagner S, Chugh A, Ramos KS, Srivastava S, Higdon D, Tollerud DJ, DeFilippis A, Becher C, Wyatt B, McCracken J, Abplanalp W, Rai SN, Ciszewski T, Xie Z, Yeager R, Prabhu SD, Bhatnagar A. Acrolein exposure is associated with increased cardiovascular disease risk. J Am Heart Assoc 2014; 3:jah3635. [PMID: 25099132 PMCID: PMC4310380 DOI: 10.1161/jaha.114.000934] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Acrolein is a reactive aldehyde present in high amounts in coal, wood, paper, and tobacco smoke. It is also generated endogenously by lipid peroxidation and the oxidation of amino acids by myeloperoxidase. In animals, acrolein exposure is associated with the suppression of circulating progenitor cells and increases in thrombosis and atherogenesis. The purpose of this study was to determine whether acrolein exposure in humans is also associated with increased cardiovascular disease (CVD) risk. Methods and Results Acrolein exposure was assessed in 211 participants of the Louisville Healthy Heart Study with moderate to high (CVD) risk by measuring the urinary levels of the major acrolein metabolite—3‐hydroxypropylmercapturic acid (3‐HPMA). Generalized linear models were used to assess the association between acrolein exposure and parameters of CVD risk, and adjusted for potential demographic confounders. Urinary 3‐HPMA levels were higher in smokers than nonsmokers and were positively correlated with urinary cotinine levels. Urinary 3‐HPMA levels were inversely related to levels of both early (AC133+) and late (AC133−) circulating angiogenic cells. In smokers as well as nonsmokers, 3‐HPMA levels were positively associated with both increased levels of platelet–leukocyte aggregates and the Framingham Risk Score. No association was observed between 3‐HPMA and plasma fibrinogen. Levels of C‐reactive protein were associated with 3‐HPMA levels in nonsmokers only. Conclusions Regardless of its source, acrolein exposure is associated with platelet activation and suppression of circulating angiogenic cell levels, as well as increased CVD risk.
Collapse
Affiliation(s)
- Natasha DeJarnett
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Department of Environmental and Occupational Health Sciences, University of Louisville, Louisville, KY (N.D.J., D.J.T., R.Y.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Daniel J Conklin
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Daniel W Riggs
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - John A Myers
- Department of Pediatrics, University of Louisville, Louisville, KY (J.A.M.)
| | - Timothy E O'Toole
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Ihab Hamzeh
- Baylor College of Medicine, Houston, TX (I.H.)
| | - Stephen Wagner
- Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Atul Chugh
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Kenneth S Ramos
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, KY (K.S.R., A.B.)
| | - Sanjay Srivastava
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Deirdre Higdon
- Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - David J Tollerud
- Department of Environmental and Occupational Health Sciences, University of Louisville, Louisville, KY (N.D.J., D.J.T., R.Y.)
| | - Andrew DeFilippis
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.) Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.F.)
| | - Carrie Becher
- Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Brad Wyatt
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - James McCracken
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Wes Abplanalp
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Shesh N Rai
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Department of Bioinformatics and Biostatics, University of Louisville, Louisville, KY (S.N.R.) Biostatistics Shared Facility, JG Brown Cancer Center, University of Louisville, Louisville, KY (S.N.R.)
| | - Tiffany Ciszewski
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Zhengzhi Xie
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Ray Yeager
- Department of Environmental and Occupational Health Sciences, University of Louisville, Louisville, KY (N.D.J., D.J.T., R.Y.)
| | - Sumanth D Prabhu
- Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.) Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL (S.D.P.)
| | - Aruni Bhatnagar
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, KY (K.S.R., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| |
Collapse
|
696
|
Blaha MJ, Silverman MG, Budoff MJ. Is there a role for coronary artery calcium scoring for management of asymptomatic patients at risk for coronary artery disease?: Clinical risk scores are not sufficient to define primary prevention treatment strategies among asymptomatic patients. Circ Cardiovasc Imaging 2014; 7:398-408; discussion 408. [PMID: 24642922 DOI: 10.1161/circimaging.113.000341] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Although risk factors have proven to be useful therapeutic targets, they are poor predictors of risk. Traditional risk scores are moderately successful in predicting future CHD events and can be a starting place for general risk categorization. However, there is substantial heterogeneity between traditional risk and actual atherosclerosis burden, with event rates predominantly driven by burden of atherosclerosis. Serum biomarkers have yet to show any clinically significant incremental value to the FRS and even when combined cannot match the predictive value of atherosclerosis imaging. As clinicians, are we willing to base therapy decisions on risk models that lack optimum-achievable accuracy and limit personalization? The decision to treat a patient in primary prevention must be a careful one because the benefit of therapy in an asymptomatic patient must clearly outweigh the potential risk. CAC, in particular, provides a personalized assessment of risk and may identify patients who will be expected to derive the most, and the least, net absolute benefit from treatment. Emerging evidence hints that CAC may also promote long-term adherence to aspirin, exercise, diet, and statin therapy. When potentially lifelong treatment decisions are on the line, clinicians must arm their patients with the most accurate risk prediction tools, and subclinical atherosclerosis testing with CAC is, at the present time, superior to any combination of risk factors and serum biomarkers.
Collapse
Affiliation(s)
- Michael J Blaha
- The Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD
| | | | | |
Collapse
|
697
|
Hochholzer W, Valina CM, Stratz C, Amann M, Schlittenhardt D, Büttner HJ, Trenk D, Neumann FJ. High-sensitivity cardiac troponin for risk prediction in patients with and without coronary heart disease. Int J Cardiol 2014; 176:444-9. [PMID: 25127973 DOI: 10.1016/j.ijcard.2014.07.094] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 05/30/2014] [Accepted: 07/26/2014] [Indexed: 01/08/2023]
Abstract
BACKGROUND In stable patients with unknown coronary anatomy, higher levels of cardiac troponin are associated with an increased risk of cardiovascular events. It was supposed that this association might be explained by the ability of cardiac troponin to detect minor myocardial necrosis which might be caused by subclinical coronary atherosclerosis. Thus, this analysis tested if the predictive value of high-sensitivity troponin T (hsTnT) in stable patients is dependent of the presence or absence of angiographically documented coronary heart disease. METHODS Stable patients undergoing elective coronary angiography were enrolled (n=2046). HsTnT was determined before diagnostic procedures. The patients were followed for up to seven years. Primary endpoint was all-cause mortality or non-fatal myocardial infarction. All endpoints were adjudicated by independent physicians. Results were adjusted to a clinical model including independent clinical predictors of the primary endpoint. RESULTS Out of the 2046 patients enrolled, 1236 (60%) had a diagnosis of obstructive coronary heart disease. HsTnT predicted independently the primary endpoint (adjusted HR 1.33, 95%-CI 1.21-1.46, P<0.001). The use of hsTnT in addition to the clinical model significantly improved discrimination (c-statistic: 0.751 to 0.773, P<0.001) as well as reclassification of the primary endpoint (NRI=0.362, P<0.001). This significant improvement persisted across various subsets and was independent of the presence of clinically detectable coronary heart disease and other variables. CONCLUSION The use of hsTnT in addition to clinical variables significantly improves discrimination and reclassification of patients with respect to all-cause mortality or non-fatal myocardial infarction irrespective of the presence of clinically detectable coronary heart disease. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov (Identifier: NCT00457236).
Collapse
Affiliation(s)
- Willibald Hochholzer
- Universitaets-Herzzentrum Freiburg·Bad Krozingen, Klinik für Kardiologie und Angiologie II, Suedring 15, 79189 Bad Krozingen, Germany.
| | - Christian M Valina
- Universitaets-Herzzentrum Freiburg·Bad Krozingen, Klinik für Kardiologie und Angiologie II, Suedring 15, 79189 Bad Krozingen, Germany
| | - Christian Stratz
- Universitaets-Herzzentrum Freiburg·Bad Krozingen, Klinik für Kardiologie und Angiologie II, Suedring 15, 79189 Bad Krozingen, Germany
| | - Michael Amann
- Universitaets-Herzzentrum Freiburg·Bad Krozingen, Klinik für Kardiologie und Angiologie II, Suedring 15, 79189 Bad Krozingen, Germany
| | - Daniel Schlittenhardt
- Universitaets-Herzzentrum Freiburg·Bad Krozingen, Klinik für Kardiologie und Angiologie II, Suedring 15, 79189 Bad Krozingen, Germany
| | - Heinz Joachim Büttner
- Universitaets-Herzzentrum Freiburg·Bad Krozingen, Klinik für Kardiologie und Angiologie II, Suedring 15, 79189 Bad Krozingen, Germany
| | - Dietmar Trenk
- Universitaets-Herzzentrum Freiburg·Bad Krozingen, Klinik für Kardiologie und Angiologie II, Suedring 15, 79189 Bad Krozingen, Germany
| | - Franz-Josef Neumann
- Universitaets-Herzzentrum Freiburg·Bad Krozingen, Klinik für Kardiologie und Angiologie II, Suedring 15, 79189 Bad Krozingen, Germany
| |
Collapse
|
698
|
Matsushita K, Sang Y, Ballew SH, Astor BC, Hoogeveen RC, Solomon SD, Ballantyne CM, Woodward M, Coresh J. Cardiac and kidney markers for cardiovascular prediction in individuals with chronic kidney disease: the Atherosclerosis Risk in Communities study. Arterioscler Thromb Vasc Biol 2014; 34:1770-7. [PMID: 24876355 PMCID: PMC4172337 DOI: 10.1161/atvbaha.114.303465] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 05/15/2014] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Traditional predictors suboptimally predict cardiovascular disease (CVD) in individuals with chronic kidney disease (CKD). This study compared 5 nontraditional cardiac and kidney markers on the improvement of cardiovascular prediction among those with CKD. APPROACH AND RESULTS Among 8622 participants aged 52 to 75 years in the Atherosclerosis Risk in Communities (ARIC) Study, cardiac troponin T, N-terminal pro-B-type natriuretic peptide, cystatin C, β2-microglobulin, and β-trace protein were compared for improvement in predicting incident CVD after stratifying by CKD status (940 participants with CKD [kidney dysfunction or albuminuria]). During a median follow-up of 11.9 years, there were 1672 CVD events including coronary disease, stroke, and heart failure (336 cases in CKD). Every marker was independently associated with incident CVD in participants with and without CKD. The adjusted hazard ratios (per 1 SD) were larger for cardiac markers than for kidney markers, particularly in CKD (1.61 [95% confidence interval, 1.43-1.81] for cardiac troponin T, 1.50 [1.34-1.68] for N-terminal pro-B-type natriuretic peptide, and <1.26 for kidney markers). Particularly in CKD group, cardiac markers compared with kidney markers contributed to greater c-statistic increment (0.032-0.036 versus 0.012-0.015 from 0.679 with only conventional predictors in CKD and 0.008-0.011 versus 0.002-0.010 from 0.697 in non-CKD) and categorical net reclassification improvement (0.086-0.127 versus 0.020-0.066 in CKD and 0.057-0.077 versus 0.014-0.048 in non-CKD). The superiority of cardiac markers was largely consistent in individual CVD outcomes. CONCLUSIONS A greater improvement in cardiovascular prediction was observed for cardiac markers than for kidney markers in people with CKD. These results suggest that cardiac troponin T and N-terminal pro-B-type natriuretic peptide are useful for better CVD risk classification in this population.
Collapse
Affiliation(s)
- Kunihiro Matsushita
- From the Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M., Y.S., S.H.B., M.W., J.C.); Department of Medicine and Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison (B.C.A.); Department of Medicine, Section of Atherosclerosis and Vascular Medicine, Baylor College of Medicine, and the Methodist DeBakey Heart and Vascular Center, Houston, TX (R.C.H., C.M.B.); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.D.S.).
| | - Yingying Sang
- From the Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M., Y.S., S.H.B., M.W., J.C.); Department of Medicine and Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison (B.C.A.); Department of Medicine, Section of Atherosclerosis and Vascular Medicine, Baylor College of Medicine, and the Methodist DeBakey Heart and Vascular Center, Houston, TX (R.C.H., C.M.B.); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.D.S.)
| | - Shoshana H Ballew
- From the Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M., Y.S., S.H.B., M.W., J.C.); Department of Medicine and Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison (B.C.A.); Department of Medicine, Section of Atherosclerosis and Vascular Medicine, Baylor College of Medicine, and the Methodist DeBakey Heart and Vascular Center, Houston, TX (R.C.H., C.M.B.); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.D.S.)
| | - Brad C Astor
- From the Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M., Y.S., S.H.B., M.W., J.C.); Department of Medicine and Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison (B.C.A.); Department of Medicine, Section of Atherosclerosis and Vascular Medicine, Baylor College of Medicine, and the Methodist DeBakey Heart and Vascular Center, Houston, TX (R.C.H., C.M.B.); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.D.S.)
| | - Ron C Hoogeveen
- From the Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M., Y.S., S.H.B., M.W., J.C.); Department of Medicine and Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison (B.C.A.); Department of Medicine, Section of Atherosclerosis and Vascular Medicine, Baylor College of Medicine, and the Methodist DeBakey Heart and Vascular Center, Houston, TX (R.C.H., C.M.B.); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.D.S.)
| | - Scott D Solomon
- From the Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M., Y.S., S.H.B., M.W., J.C.); Department of Medicine and Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison (B.C.A.); Department of Medicine, Section of Atherosclerosis and Vascular Medicine, Baylor College of Medicine, and the Methodist DeBakey Heart and Vascular Center, Houston, TX (R.C.H., C.M.B.); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.D.S.)
| | - Christie M Ballantyne
- From the Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M., Y.S., S.H.B., M.W., J.C.); Department of Medicine and Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison (B.C.A.); Department of Medicine, Section of Atherosclerosis and Vascular Medicine, Baylor College of Medicine, and the Methodist DeBakey Heart and Vascular Center, Houston, TX (R.C.H., C.M.B.); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.D.S.)
| | - Mark Woodward
- From the Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M., Y.S., S.H.B., M.W., J.C.); Department of Medicine and Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison (B.C.A.); Department of Medicine, Section of Atherosclerosis and Vascular Medicine, Baylor College of Medicine, and the Methodist DeBakey Heart and Vascular Center, Houston, TX (R.C.H., C.M.B.); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.D.S.)
| | - Josef Coresh
- From the Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M., Y.S., S.H.B., M.W., J.C.); Department of Medicine and Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison (B.C.A.); Department of Medicine, Section of Atherosclerosis and Vascular Medicine, Baylor College of Medicine, and the Methodist DeBakey Heart and Vascular Center, Houston, TX (R.C.H., C.M.B.); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.D.S.)
| |
Collapse
|
699
|
Postinjury hyperfibrinogenemia compromises efficacy of heparin-based venous thromboembolism prophylaxis. Shock 2014; 41:33-9. [PMID: 24351527 DOI: 10.1097/shk.0000000000000067] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND Venous thromboembolism (VTE) prophylaxis remains debated following trauma, and recommendations have not been established. Although hyperfibrinogenemia is a marker of proinflammatory states, it also contributes to thrombus formation. Postinjury hyperfibrinogenemia is common, but the effect of hyperfibrinogenemia on VTE prophylaxis has not been fully elucidated. Therefore, we hypothesized that heparin is less effective for VTE prophylaxis following severe injury due to hyperfibrinogenemia. METHODS In vitro studies evaluated thromboelastography (TEG) parameters in 10 healthy volunteers after the addition of fibrinogen concentrate and heparin. Data from a recent randomized controlled trial, conducted at an academic level I trauma center surgical intensive care unit, were reviewed. Critically injured patients were randomized to standard VTE prophylaxis (5,000 U low-molecular-weight heparin daily) or TEG-guided prophylaxis (up to 10,000 U low-molecular-weight heparin daily) and were followed up for 5 days. Analysis was performed to evaluate the relationship between fibrinogen levels, measures of anticoagulation, and TEG parameters. RESULTS In vitro studies revealed increased fibrinogen reversed the effects of heparin as measured by TEG. Fifty patients were enrolled in the clinical study with 25 in each arm. Thromboelastography parameters, fibrinogen, platelet count, and anti-Xa levels did not differ between groups despite treatment provided. Fibrinogen levels increased over the 5-day study period (597 ± 24.0 to 689.3 ± 25.0), as well as clot strength (9.8 ± 0.4 to 14.5 ± 0.6), which had a significant correlation coefficient (P < 0.01). Moreover, there was a moderate inverse correlation between fibrinogen level and the effect of heparin (RF), which was significant on study days 1 and 3, implicating hyperfibrinogenemia in heparin resistance. CONCLUSIONS Hypercoagulability and heparin resistance are common following trauma. The preclinical and clinical relationships between fibrinogen levels and hypercoagulability implicate hyperfibrinogenemia as a potential factor in heparin resistance.
Collapse
|
700
|
Golia E, Limongelli G, Natale F, Fimiani F, Maddaloni V, Russo PE, Riegler L, Bianchi R, Crisci M, Palma GD, Golino P, Russo MG, Calabrò R, Calabrò P. Adipose tissue and vascular inflammation in coronary artery disease. World J Cardiol 2014; 6:539-554. [PMID: 25068015 PMCID: PMC4110603 DOI: 10.4330/wjc.v6.i7.539] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 03/25/2014] [Accepted: 06/03/2014] [Indexed: 02/06/2023] Open
Abstract
Obesity has become an important public health issue in Western and developing countries, with well known metabolic and cardiovascular complications. In the last decades, evidence have been growing about the active role of adipose tissue as an endocrine organ in determining these pathological consequences. As a consequence of the expansion of fat depots, in obese subjects, adipose tissue cells develope a phenotypic modification, which turns into a change of the secretory output. Adipocytokines produced by both adipocytes and adipose stromal cells are involved in the modulation of glucose and lipid handling, vascular biology and, moreover, participate to the systemic inflammatory response, which characterizes obesity and metabolic syndrome. This might represent an important pathophysiological link with atherosclerotic complications and cardiovascular events. A great number of adipocytokines have been described recently, linking inflammatory mileu and vascular pathology. The understanding of these pathways is crucial not only from a pathophysiological point of view, but also to a better cardiovascular disease risk stratification and to the identification of possible therapeutic targets. The aim of this paper is to review the role of Adipocytokines as a possible link between obesity and vascular disease.
Collapse
|