1
|
Masini G, Gargani L, Morizzo C, Guarini G, Bort IR, Baldini M, Tamborrino PP, Vitale C, Palombo C, De Caterina R. Prognostic impact of peripheral artery disease-related parameters in patients with acute coronary syndrome. J Cardiovasc Med (Hagerstown) 2024; 25:749-756. [PMID: 39101369 DOI: 10.2459/jcm.0000000000001653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
BACKGROUND Lower extremity arterial disease (LEAD) and increased aortic stiffness are associated with higher mortality in patients with chronic coronary syndrome, while their prognostic significance after an acute coronary syndrome (ACS) is less known. METHODS We analyzed prevalence, clinical phenotypes and association of LEAD - assessed by the ankle-brachial index (ABI) - and increased aortic stiffness - assessed by the aortic pulse wave velocity (PWV) - with all-cause mortality and major adverse cardiovascular events (MACE) in patients admitted with an ACS. RESULTS Among 270 patients admitted for ACS (mean age 67 years, 80% males), 41 (15%) had an ABI ≤0.9, with 14 of them (34%) presenting with intermittent claudication (symptomatic LEAD). Patients with symptomatic LEAD, compared with those with asymptomatic LEAD or without LEAD, had higher prevalence of cardiovascular risk factors, lower estimated glomerular filtration rate and higher high-sensitivity C-reactive protein. Patients with LEAD, either symptomatic or asymptomatic, more frequently presented with non-ST-elevation myocardial infarction and more frequently had multivessel coronary artery disease. Both symptomatic and asymptomatic LEAD were significantly associated with all-cause mortality after adjustment for confounders, including multivessel disease or carotid artery disease (hazard ratio 4.03, 95% confidence interval 1.61-10.08, P < 0.01), whereas PWV was not associated with the outcome in the univariable model. LEAD and PWV were not associated with a higher risk of MACE (myocardial infarction or unstable angina, stroke, or transient ischemic attack). CONCLUSIONS LEAD, either clinical or subclinical, but not increased aortic stiffness, is an independent predictor of all-cause mortality in patients admitted for ACS.
Collapse
Affiliation(s)
- Gabriele Masini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, Pisa University Hospital, and Chair of Cardiology, University of Pisa, Pisa, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Oflar E, Akdeniz E, Yıldız C, Koyuncu A, Mavi B, Karabulut D, Çağlar FNT, Kavala AA, Türkyılmaz S. Evaluation of systemic immune-inflammation index for predicting severity of lower extremity arterial disease. Vascular 2024; 32:797-803. [PMID: 38705727 DOI: 10.1177/17085381241251772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
OBJECTIVE Predictive value of systemic immune-inflammation index (SII) has been shown in clinical outcomes and complexity of coronary artery disease, acute coronary syndrome, and heart failure. We sight to evaluate value of SII in patients with lower extremity arterial disease (LEAD). METHODS A total of 271 patients diagnosed with LEAD were included to our study. Blood samples of the patients were collected and analyzed for biochemical variables and complete blood count parameters. SII value of each patient was calculated. The complexity of atherosclerotic disease was classified according to Trans-Atlantic Inter-Society Consensus (TASC II) classification. RESULTS Patients with TASC C-D were older than patients in TASC A-B group (63.06 ± 9.24 years and 60.85 ± 8.75 years, respectively). Other co-morbidities were comparable in both groups. Hemoglobin level and lymphocyte count were significantly lower, neutrophil, platelet counts, and SII values were significantly higher in patients with TASC C-D disease compared to that of patients with TASC A-B disease. SII showed significant correlation with the severity of LEAD (r = 0.363, p < .001). SII value of 664.24 predicted TASC C-D disease with a sensitivity and specificity of 60.8% and 73.3%, respectively. Results of multivariate logistic regression analysis showed that SII had higher odds ratio compared to platelet, neutrophil, and lymphocyte counts. CONCLUSION Higher SII may indicate probability of more complex LEAD. This relationship seems plausible in terms of similar pathophysiology of coronary artery disease and peripheral artery disease.
Collapse
Affiliation(s)
- Ersan Oflar
- Cardiology Department, Bakırkoy Dr Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Evliya Akdeniz
- Cardiology Department, Bakırkoy Dr Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Cennet Yıldız
- Cardiology Department, Bakırkoy Dr Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Atilla Koyuncu
- Cardiology Department, Bakırkoy Dr Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Büşra Mavi
- Cardiology Department, Bakırkoy Dr Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Dilay Karabulut
- Cardiology Department, Bakırkoy Dr Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | | | - Ali Aycan Kavala
- Cardiovascular Surgery Department, Bakırkoy Dr Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Saygın Türkyılmaz
- Cardiovascular Surgery Department, Bakırkoy Dr Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
3
|
Fantini DA, Yang G, Khanna A, Subramanian D, Phillippi JA, Huang NF. Overcoming big bottlenecks in vascular regeneration. Commun Biol 2024; 7:876. [PMID: 39020071 PMCID: PMC11255241 DOI: 10.1038/s42003-024-06567-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 07/05/2024] [Indexed: 07/19/2024] Open
Abstract
Bioengineering and regenerative medicine strategies are promising for the treatment of vascular diseases. However, current limitations inhibit the ability of these approaches to be translated to clinical practice. Here we summarize some of the big bottlenecks that inhibit vascular regeneration in the disease applications of aortic aneurysms, stroke, and peripheral artery disease. We also describe the bottlenecks preventing three-dimensional bioprinting of vascular networks for tissue engineering applications. Finally, we describe emerging technologies and opportunities to overcome these challenges to advance vascular regeneration.
Collapse
Affiliation(s)
- Dalia A Fantini
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Guang Yang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
- Epicrispr Biotechnologies, Inc, South San Francisco, CA, USA
| | | | - Divya Subramanian
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Julie A Phillippi
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Ngan F Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford, CA, USA.
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA.
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
4
|
Wang X, Nai YH, Gan J, Lian CPL, Ryan FK, Tan FSL, Chan DYS, Ng JJ, Lo ZJ, Chong TT, Hausenloy DJ. Multi-Modality Imaging of Atheromatous Plaques in Peripheral Arterial Disease: Integrating Molecular and Imaging Markers. Int J Mol Sci 2023; 24:11123. [PMID: 37446302 DOI: 10.3390/ijms241311123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/14/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Peripheral artery disease (PAD) is a common and debilitating condition characterized by the narrowing of the limb arteries, primarily due to atherosclerosis. Non-invasive multi-modality imaging approaches using computed tomography (CT), magnetic resonance imaging (MRI), and nuclear imaging have emerged as valuable tools for assessing PAD atheromatous plaques and vessel walls. This review provides an overview of these different imaging techniques, their advantages, limitations, and recent advancements. In addition, this review highlights the importance of molecular markers, including those related to inflammation, endothelial dysfunction, and oxidative stress, in PAD pathophysiology. The potential of integrating molecular and imaging markers for an improved understanding of PAD is also discussed. Despite the promise of this integrative approach, there remain several challenges, including technical limitations in imaging modalities and the need for novel molecular marker discovery and validation. Addressing these challenges and embracing future directions in the field will be essential for maximizing the potential of molecular and imaging markers for improving PAD patient outcomes.
Collapse
Affiliation(s)
- Xiaomeng Wang
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Ying-Hwey Nai
- Clinical Imaging Research Centre, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Julian Gan
- Siemens Healthineers, Singapore 348615, Singapore
| | - Cheryl Pei Ling Lian
- Health and Social Sciences Cluster, Singapore Institute of Technology, Singapore 138683, Singapore
| | - Fraser Kirwan Ryan
- Infocomm Technology Cluster, Singapore Institute of Technology, Singapore 138683, Singapore
| | - Forest Su Lim Tan
- Infocomm Technology Cluster, Singapore Institute of Technology, Singapore 138683, Singapore
| | - Dexter Yak Seng Chan
- Department of General Surgery, Khoo Teck Puat Hospital, Singapore 768828, Singapore
| | - Jun Jie Ng
- Division of Vascular and Endovascular Surgery, Department of Cardiac, Thoracic and Vascular Surgery, National University Heart Centre, Singapore 119074, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Zhiwen Joseph Lo
- Vascular Surgery Service, Department of Surgery, Woodlands Health, Singapore 258499, Singapore
- Centre for Population Health Sciences, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Tze Tec Chong
- Department of Vascular Surgery, Singapore General Hospital, Singapore 168752, Singapore
- Surgical Academic Clinical Programme, Singapore General Hospital, Singapore 169608, Singapore
- Vascular SingHealth Duke-NUS Disease Centre, Singapore 168752, Singapore
| | - Derek John Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
- Yong Loo Lin School of Medicine, National University Singapore, Singapore 117597, Singapore
- The Hatter Cardiovascular Institute, University College London, London WC1E 6HX, UK
| |
Collapse
|
5
|
Zamzam A, Syed MH, Rotstein OD, Eikelboom J, Klein DJ, Singh KK, Abdin R, Qadura M. Validating fatty acid binding protein 3 as a diagnostic and prognostic biomarker for peripheral arterial disease: A three-year prospective follow-up study. EClinicalMedicine 2023; 55:101766. [PMID: 36531981 PMCID: PMC9755058 DOI: 10.1016/j.eclinm.2022.101766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Patients with peripheral arterial disease (PAD) often remain undiagnosed and therefore suboptimally managed. Here, we investigated the diagnostic and prognostic potential of fatty acid binding protein 3 (FABP3) in patients with PAD. METHODS In the discovery phase, 374 PAD and 184 non-PAD patients were recruited from vascular surgery ambulatory clinics at St. Michael's Hospital (Toronto, Ontario, Canada) between October 4, 2017 to October 29, 2018. The diagnostic ability of baseline FABP3 level was investigated through receiver operator characteristic (ROC) curves to determine two cutoff points: 1) an exclusionary "rule out" cutoff point, and 2) a confirmatory "rule in" cutoff point. Next, these cutoff points were confirmed in the external validation phase using a separate cohort of 312 patients (180 PAD and 132 non-PAD) recruited from ambulatory vascular surgery clinics at St. Michael's Hospital (Canada) between November 6, 2018-July 30, 2019. Cox regression analyses were used to explore the independent association between FABP3 and major adverse limb events (MALE - defined as need for arterial revascularization or major amputation) and decrease in ankle-brachial index (ABI -defined as drop ≥0.15) during 3 years of follow-up. FINDINGS In the discovery phase, FABP3 levels were significantly elevated in patients with PAD compared to non-PAD patients. ROC analysis demonstrated that FABP3 had an AUC of 0.83 (95% CI: 0.81-0.86, p-value < 0.001). FABP3 exclusionary cutoff was <1.55 ng/ml (sensitivity = 96%; specificity = 40%), whereas FABP3 confirmatory cutoff was >3.55 ng/ml (sensitivity = 43%; specificity = 95%) - values that were confirmed in the external validation phase. Cox regression analysis demonstrated FABP3 to be an independent predictor of increase in MALE [HR = 1.14 (1.03-1.29); p-value = 0.010] and worsening PAD status (drop in ABI >0.15 [HR = 1.11 (1.02-1.19); p-value = 0.009]). INTERPRETATION Our findings suggested that FABP3 levels can be used as both a diagnostic and prognostic biomarker for PAD, and may facilitate risk stratification in select individuals for purposes of vascular evaluation or intensive medical management. FUNDING Funding for this study was provided by the Bill and Vicky Blair Foundation.
Collapse
Affiliation(s)
- Abdelrahman Zamzam
- Division of Vascular Surgery, St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
| | - Muzammil H. Syed
- Division of Vascular Surgery, St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
| | - Ori D. Rotstein
- Department of Surgery, University of Toronto, Toronto, ON, M5S 1A1, Canada
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
| | - John Eikelboom
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, ON, L8S 4K1, Canada
| | - David J. Klein
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
- Department of Critical Care, St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
| | - Krishna K. Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London ON, N6A 5C1, Canada
| | - Rawand Abdin
- Department of Medicine, McMaster University, Hamilton, ON, L8S 4K1, Canada
| | - Mohammad Qadura
- Division of Vascular Surgery, St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
- Department of Surgery, University of Toronto, Toronto, ON, M5S 1A1, Canada
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
- Corresponding author. St. Michael's Hospital, 30 Bond St, 7-076 Bond Wing, Toronto, Ontario, M5B 1W8, Canada.
| |
Collapse
|
6
|
Ziegler L, Hedin U, Gottsäter A. Circulating Biomarkers in Lower Extremity Artery Disease. Eur Cardiol 2022; 17:e09. [PMID: 35401792 PMCID: PMC8978021 DOI: 10.15420/ecr.2021.58] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/31/2022] [Indexed: 12/24/2022] Open
Abstract
Lower extremity artery disease (LEAD), a chronic condition with disturbed lower extremity circulation due to narrowing of the arteries, is predominantly caused by atherosclerosis and is associated with the presence of cardiovascular risk factors and an increased risk of cardiovascular events. LEAD is prevalent among older individuals and predicted to rise with the ageing population. In progressive disease, the patient experiences symptoms of ischaemia when walking and, in advanced critical limb-threatening ischaemia, even at rest. However, LEAD is asymptomatic in most patients, delaying diagnosis and treatment. In this setting, circulating biomarkers may facilitate earlier diagnosis in selected individuals. This review provides a broad overview of the circulating biomarkers investigated to date in relation to LEAD and discusses their usefulness in clinical practice.
Collapse
Affiliation(s)
- Louise Ziegler
- Division of Internal Medicine, Department of Clinical Sciences, Karolinska Institute, Danderyd Hospital, Stockholm, Sweden
| | - Ulf Hedin
- Vascular Surgery Division, Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Anders Gottsäter
- Department of Medicine, Lund University, Malmö, Sweden; Department of Medicine, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
7
|
Dinoto E, Ferlito F, La Marca MA, Tortomasi G, Urso F, Evola S, Guercio G, Marcianò M, Pakeliani D, Bajardi G, Pecoraro F. The Role of Early Revascularization and Biomarkers in the Management of Diabetic Foot Ulcers: A Single Center Experience. Diagnostics (Basel) 2022; 12:diagnostics12020538. [PMID: 35204630 PMCID: PMC8871223 DOI: 10.3390/diagnostics12020538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/08/2022] [Accepted: 02/16/2022] [Indexed: 11/23/2022] Open
Abstract
Diabetic neuropathy and Peripheral Arterial Disease (PAD) are the main etiological factors in foot ulceration. Herein, we report our experience of diabetic foot ulceration (DFU) management, with an analysis of the relationship between the rate of lower extremity amputation, in persons with infected DFU, after revascularization procedures performed to prevent major amputation. This study highlights the role of different biomarkers, showing their usefulness and potentiality in diabetic foot ulcer management, especially for the early diagnosis and therapy effectiveness monitoring. A retrospective analysis, from September 2016 to January 2021, of diabetic patients presenting diabetic foot with DFU, was performed. All patients were treated with at least one vascular procedure (endovascular, open, hybrid procedures) targeting PAD lesions. Outcomes measured were perioperative mortality and morbidity. Freedom from occlusion, primary and secondary patency, and amputation rate were registered. A total of 267 patients, with a mean age of 72.5 years, were included in the study. The major amputation rate was 6.2%, minor amputation rate was 17%. In our experience, extreme revascularization to obtain direct flow reduced the rate of amputations, with an increase in ulcer healing.
Collapse
Affiliation(s)
- Ettore Dinoto
- Vascular Surgery Unit, AOUP Policlinico “P. Giaccone”, 90127 Palermo, Italy; (F.F.); (M.A.L.M.); (G.T.); (F.U.); (G.B.); (F.P.)
- Correspondence:
| | - Francesca Ferlito
- Vascular Surgery Unit, AOUP Policlinico “P. Giaccone”, 90127 Palermo, Italy; (F.F.); (M.A.L.M.); (G.T.); (F.U.); (G.B.); (F.P.)
| | - Manfredi Agostino La Marca
- Vascular Surgery Unit, AOUP Policlinico “P. Giaccone”, 90127 Palermo, Italy; (F.F.); (M.A.L.M.); (G.T.); (F.U.); (G.B.); (F.P.)
| | - Graziella Tortomasi
- Vascular Surgery Unit, AOUP Policlinico “P. Giaccone”, 90127 Palermo, Italy; (F.F.); (M.A.L.M.); (G.T.); (F.U.); (G.B.); (F.P.)
| | - Francesca Urso
- Vascular Surgery Unit, AOUP Policlinico “P. Giaccone”, 90127 Palermo, Italy; (F.F.); (M.A.L.M.); (G.T.); (F.U.); (G.B.); (F.P.)
| | - Salvatore Evola
- Unit of Cardiology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE) “G. D’Alessandro”, University Hospital Paolo Giaccone, University of Palermo, 90127 Palermo, Italy;
| | - Giovanni Guercio
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy;
- Department of General and Emergency Surgery, Paolo Giaccone University Hospital, 90127 Palermo, Italy;
| | - Marco Marcianò
- Department of General and Emergency Surgery, Paolo Giaccone University Hospital, 90127 Palermo, Italy;
| | - David Pakeliani
- Vascular Surgery Unit, Ospedali Riuniti Villa Sofia-Cervello, 90100 Palermo, Italy;
| | - Guido Bajardi
- Vascular Surgery Unit, AOUP Policlinico “P. Giaccone”, 90127 Palermo, Italy; (F.F.); (M.A.L.M.); (G.T.); (F.U.); (G.B.); (F.P.)
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy;
| | - Felice Pecoraro
- Vascular Surgery Unit, AOUP Policlinico “P. Giaccone”, 90127 Palermo, Italy; (F.F.); (M.A.L.M.); (G.T.); (F.U.); (G.B.); (F.P.)
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy;
| |
Collapse
|
8
|
X‑irradiation induces acute and early term inflammatory responses in atherosclerosis‑prone ApoE‑/‑ mice and in endothelial cells. Mol Med Rep 2021; 23:399. [PMID: 33786610 PMCID: PMC8025474 DOI: 10.3892/mmr.2021.12038] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/09/2020] [Indexed: 01/09/2023] Open
Abstract
Thoracic radiotherapy is an effective treatment for many types of cancer; however it is also associated with an increased risk of developing cardiovascular disease (CVD), appearing mainly ≥10 years after radiation exposure. The present study investigated acute and early term physiological and molecular changes in the cardiovascular system after ionizing radiation exposure. Female and male ApoE‑/‑ mice received a single exposure of low or high dose X‑ray thoracic irradiation (0.1 and 10 Gy). The level of cholesterol and triglycerides, as well as a large panel of inflammatory markers, were analyzed in serum samples obtained at 24 h and 1 month after irradiation. The secretion of inflammatory markers was further verified in vitro in coronary artery and microvascular endothelial cell lines after exposure to low and high dose of ionizing radiation (0.1 and 5 Gy). Local thoracic irradiation of ApoE‑/‑ mice increased serum growth differentiation factor‑15 (GDF‑15) and C‑X‑C motif chemokine ligand 10 (CXCL10) levels in both female and male mice 24 h after high dose irradiation, which were also secreted from coronary artery and microvascular endothelial cells in vitro. Sex‑specific responses were observed for triglyceride and cholesterol levels, and some of the assessed inflammatory markers as detailed below. Male ApoE‑/‑ mice demonstrated elevated intercellular adhesion molecule‑1 and P‑selectin at 24 h, and adiponectin and plasminogen activator inhibitor‑1 at 1 month after irradiation, while female ApoE‑/‑ mice exhibited decreased monocyte chemoattractant protein‑1 and urokinase‑type plasminogen activator receptor at 24 h, and basic fibroblast growth factor 1 month after irradiation. The inflammatory responses were mainly significant following high dose irradiation, but certain markers showed significant changes after low dose exposure. The present study revealed that acute/early inflammatory responses occurred after low and high dose thoracic irradiation. However, further research is required to elucidate early asymptomatic changes in the cardiovascular system post thoracic X‑irradiation and to investigate whether GDF‑15 and CXCL10 could be considered as potential biomarkers for the early detection of CVD risk in thoracic radiotherapy‑treated patients.
Collapse
|
9
|
Saenz-Pipaon G, Martinez-Aguilar E, Orbe J, González Miqueo A, Fernandez-Alonso L, Paramo JA, Roncal C. The Role of Circulating Biomarkers in Peripheral Arterial Disease. Int J Mol Sci 2021; 22:ijms22073601. [PMID: 33808453 PMCID: PMC8036489 DOI: 10.3390/ijms22073601] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/18/2022] Open
Abstract
Peripheral arterial disease (PAD) of the lower extremities is a chronic illness predominantly of atherosclerotic aetiology, associated to traditional cardiovascular (CV) risk factors. It is one of the most prevalent CV conditions worldwide in subjects >65 years, estimated to increase greatly with the aging of the population, becoming a severe socioeconomic problem in the future. The narrowing and thrombotic occlusion of the lower limb arteries impairs the walking function as the disease progresses, increasing the risk of CV events (myocardial infarction and stroke), amputation and death. Despite its poor prognosis, PAD patients are scarcely identified until the disease is advanced, highlighting the need for reliable biomarkers for PAD patient stratification, that might also contribute to define more personalized medical treatments. In this review, we will discuss the usefulness of inflammatory molecules, matrix metalloproteinases (MMPs), and cardiac damage markers, as well as novel components of the liquid biopsy, extracellular vesicles (EVs), and non-coding RNAs for lower limb PAD identification, stratification, and outcome assessment. We will also explore the potential of machine learning methods to build prediction models to refine PAD assessment. In this line, the usefulness of multimarker approaches to evaluate this complex multifactorial disease will be also discussed.
Collapse
Affiliation(s)
- Goren Saenz-Pipaon
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, 31008 Pamplona, Spain; (G.S.-P.); (J.O.); (J.A.P.)
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain; (E.M.-A.); (A.G.M.); (L.F.-A.)
| | - Esther Martinez-Aguilar
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain; (E.M.-A.); (A.G.M.); (L.F.-A.)
- Departamento de Angiología y Cirugía Vascular, Complejo Hospitalario de Navarra, 31008 Pamplona, Spain
| | - Josune Orbe
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, 31008 Pamplona, Spain; (G.S.-P.); (J.O.); (J.A.P.)
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain; (E.M.-A.); (A.G.M.); (L.F.-A.)
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Arantxa González Miqueo
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain; (E.M.-A.); (A.G.M.); (L.F.-A.)
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Laboratory of Heart Failure, Program of Cardiovascular Diseases, Cima Universidad de Navarra, 31008 Pamplona, Spain
| | - Leopoldo Fernandez-Alonso
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain; (E.M.-A.); (A.G.M.); (L.F.-A.)
- Departamento de Angiología y Cirugía Vascular, Complejo Hospitalario de Navarra, 31008 Pamplona, Spain
| | - Jose Antonio Paramo
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, 31008 Pamplona, Spain; (G.S.-P.); (J.O.); (J.A.P.)
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain; (E.M.-A.); (A.G.M.); (L.F.-A.)
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Hematology Service, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Carmen Roncal
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, 31008 Pamplona, Spain; (G.S.-P.); (J.O.); (J.A.P.)
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain; (E.M.-A.); (A.G.M.); (L.F.-A.)
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-948194700
| |
Collapse
|
10
|
Pereira-da-Silva T, Napoleão P, Pinheiro T, Selas M, Silva F, Ferreira RC, Carmo MM. The Proinflammatory Soluble CD40 Ligand Is Associated with the Systemic Extent of Stable Atherosclerosis. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:39. [PMID: 33406736 PMCID: PMC7824733 DOI: 10.3390/medicina57010039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/26/2020] [Accepted: 12/29/2020] [Indexed: 01/05/2023]
Abstract
Background and objectives: Polyvascular atherosclerosis is frequent and associated with a high cardiovascular risk, although the mechanisms regulating the atherosclerosis extent to single or multiple arterial territories are still poorly understood. Inflammation regulates atherogenesis and soluble CD40 ligand (sCD40L) is an inflammatory mediator associated with the presence of single-territorial atherosclerosis. We assessed whether the sCD40L expression is associated with the atherosclerosis extent to single or multiple arterial territories and with the atherosclerosis severity in different territories. Materials and Methods: We prospectively enrolled 94 participants with no atherosclerosis (controls, n = 26); isolated coronary atherosclerosis (group 1, n = 20); coronary and lower extremity (LE) atherosclerosis (group 2, n = 18); coronary and carotid atherosclerosis (group 3, n = 12); and coronary, LE, and carotid atherosclerosis (group 4, n = 18). Serum sCD40L levels were quantified. Results: The sCD40L levels (ng/mL, mean (standard deviation)) were 4.0 (1.5), 5.6 (2.6), 7.2 (4.2), 5.9 (3.7), and 5.1 (2.4) in controls and groups 1 to 4, respectively (ANOVA p = 0.012). In nonrevascularized patients, the sCD40L levels were significantly higher in group 2 than in group 1 and were correlated with the number of LE diseased segments. Prior LE bypass surgery was associated with lower sCD40L levels. Coexistence of coronary and LE atherosclerosis was independently associated with the sCD40L levels. Conclusions: The sCD40L levels were increased in stable atherosclerosis, particularly in polyvascular coronary and LE atherosclerosis. The number of LE diseased segments and prior LE revascularization were associated with sCD40L expression. To our knowledge, these are novel data, which provide insights into the mechanisms underlying multi-territorial atherosclerosis expression. sCD40L may be a promising noninvasive tool for refining the stratification of the systemic atherosclerotic burden.
Collapse
Affiliation(s)
- Tiago Pereira-da-Silva
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
| | - Patrícia Napoleão
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal;
| | - Teresa Pinheiro
- Instituto de Bioengenharia e Biociências, Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Lisbon, Portugal;
| | - Mafalda Selas
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
| | - Filipa Silva
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
| | - Rui Cruz Ferreira
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
| | - Miguel Mota Carmo
- Chronic Diseases Research Center (CEDOC), NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal;
| |
Collapse
|
11
|
Soluble CD40 ligand expression in stable atherosclerosis: A systematic review and meta-analysis. Atherosclerosis 2020; 319:86-100. [PMID: 33494009 DOI: 10.1016/j.atherosclerosis.2020.12.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 11/08/2020] [Accepted: 12/11/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND AIMS The role of inflammation in atherosclerosis development and expression in different arterial territories is unclear. Soluble CD40 ligand (sCD40L) mediates inflammation and atherogenesis. Through a systematic review and meta-analysis, we assessed whether sCD40L was dysregulated in stable atherosclerosis, irrespective of the diseased arterial territory, and whether this dysregulation differed according to the specific territory. METHODS Systematic literature searches were performed in MEDLINE, Cochrane Library, Web of Science, and Embase for studies reporting circulating sCD40L levels in individuals with and without stable atherosclerosis. sCD40L levels were compared using random-effects meta-analysis, weighted by the inverse variance method (study protocol: PROSPERO CRD42020181392). RESULTS Fifty-four studies (59 estimates) including 7705 patients and 7841 controls were analyzed. sCD40L levels were found to be increased in patients with atherosclerosis, irrespective of the territory (standardized mean difference [SMD] 0.43, 95% CI 0.29-0.57; 59 estimates; χ2 heterogeneity p < 0.001; I2 = 92%). SMD was greatest in carotid atherosclerosis (SMD 0.58, 95% CI 0.30-0.86; 17 estimates), followed by coronary (SMD 0.43, 95% CI 0.24-0.62; 33 estimates), lower extremity (SMD 0.26, 95% CI -0.02-0.54; 7 estimates), and renal atherosclerosis (SMD -0.07, 95% CI -2.77-2.64; 2 estimates) (χ2 heterogeneity p < 0.001; I2 ≥ 80% for all). Subgroup analysis revealed that sCD40L levels were increased in clinical, but not subclinical, atherosclerosis. CONCLUSIONS sCD40L levels were increased in stable atherosclerosis, particularly in the carotid and coronary territories. These novel data support sCD40L as a marker of systemic atherosclerosis, possibly with differential roles in specific territories.
Collapse
|
12
|
Xu XY, Guo L, Wang Q, Yu XB, Li L, Wei Q. Association between lipoprotein-associated phospholipase A2 and lower extremity arterial disease in type 2 diabetes mellitus. Clin Chim Acta 2020; 510:228-231. [DOI: 10.1016/j.cca.2020.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 11/30/2022]
|
13
|
Refsgaard Holm M, Christensen H, Rasmussen J, Johansen ML, Schou M, Faber J, Kistorp C. Fibroblast growth factor 21 in patients with cardiac cachexia: a possible role of chronic inflammation. ESC Heart Fail 2019; 6:983-991. [PMID: 31429530 PMCID: PMC6816069 DOI: 10.1002/ehf2.12502] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 06/18/2019] [Accepted: 07/01/2019] [Indexed: 12/12/2022] Open
Abstract
Aims Cardiac cachexia is a wasting syndrome characterized by chronic inflammation and high mortality. Fibroblast growth factor 21 (FGF‐21) and monocyte chemoattractant protein 1 (MCP‐1) are associated with cardiovascular disease and systemic inflammation. We investigated FGF‐21 and MCP‐1 in relations to cardiac function, inflammation, and wasting in patients with heart failure with reduced ejection fraction (HFrEF) and cardiac cachexia. Methods and results Plasma FGF‐21 and MCP‐1 were measured in a cross‐sectional study among the three study groups: 19 patients with HFrEF with cardiac cachexia, 19 patients with HFrEF without cachexia, and 19 patients with ischaemic heart disease and preserved ejection fraction. Patients with HFrEF and cardiac cachexia displayed higher FGF‐21 levels median (inter quantile range) 381 (232–577) pg/mL than patients with HFrEF without cachexia 224 (179–309) pg/mL and ischaemic heart disease patients 221 (156–308) pg/mL (P = 0.0496). No difference in MCP‐1 levels were found among the groups (P = 0.345). In a multivariable regression analysis, FGF‐21 (logarithm 2) was independently associated with interleukin 6 (logarithm 2) (P = 0.015) and lower muscle mass (P = 0.043), while no relation with N‐terminal pro‐hormone brain natriuretic peptide was observed. Conclusions Fibroblast growth factor 21 (FGF‐21) levels were elevated in patients with HFrEF and cardiac cachexia, which could be mediated by increased inflammation and muscle wasting rather than impaired cardiac function.
Collapse
Affiliation(s)
- Maria Refsgaard Holm
- Centre of Endocrinology and Metabolism, Department of Internal Medicine, Copenhagen University Hospitals, Herlev and Gentofte, Herlev Ringvej 75, 2730, Herlev, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Heidi Christensen
- Centre of Endocrinology and Metabolism, Department of Internal Medicine, Copenhagen University Hospitals, Herlev and Gentofte, Herlev Ringvej 75, 2730, Herlev, Denmark
| | - Jon Rasmussen
- Centre of Endocrinology and Metabolism, Department of Internal Medicine, Copenhagen University Hospitals, Herlev and Gentofte, Herlev Ringvej 75, 2730, Herlev, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marie Louise Johansen
- Centre of Endocrinology and Metabolism, Department of Internal Medicine, Copenhagen University Hospitals, Herlev and Gentofte, Herlev Ringvej 75, 2730, Herlev, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten Schou
- Department of Cardiology, Copenhagen University Hospitals, Herlev and Gentofte, Herlev, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Faber
- Centre of Endocrinology and Metabolism, Department of Internal Medicine, Copenhagen University Hospitals, Herlev and Gentofte, Herlev Ringvej 75, 2730, Herlev, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Caroline Kistorp
- Department of Endocrinology, Copenhagen University Hospitals, Rigshospitalet, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Abstract
Peripheral arterial disease (PAD) is an atherosclerotic disease closely associated with high morbidity and mortality in cardiac events. Inflammation is crucial in atherosclerosis both at triggering and in progression. Numerous inflammatory biomarkers (cytokines, matrix metalloproteinases (MMPs), selectin, intracellular adhesion molecule (ICAM), vascular cell adhesion molecule (VCAM) C-reactive protein (CRP), fibrinogen) have been measured in atherosclerotic diseases including PAD. This paper summarizes the data on the inflammatory biomarkers for PAD pathophysiology and highlights the most useful markers in monitoring PAD outcomes.
Collapse
|
15
|
Li W, Dorans KS, Wilker EH, Rice MB, Ljungman PL, Schwartz JD, Coull BA, Koutrakis P, Gold DR, Keaney JF, Vasan RS, Benjamin EJ, Mittleman MA. Short-term exposure to ambient air pollution and circulating biomarkers of endothelial cell activation: The Framingham Heart Study. ENVIRONMENTAL RESEARCH 2019; 171:36-43. [PMID: 30654247 PMCID: PMC6478022 DOI: 10.1016/j.envres.2018.10.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/16/2018] [Accepted: 10/25/2018] [Indexed: 05/23/2023]
Abstract
BACKGROUND Short-term exposure to air pollution has been associated with cardiovascular events, potentially by promoting endothelial cell activation and inflammation. A few large-scale studies have examined the associations and have had mixed results. METHODS We included 3820 non-current smoking participants (mean age 56 years, 54% women) from the Framingham Offspring cohort examinations 7 (1998-2001) and 8 (2005-2008), and Third Generation cohort examination 1 (2002-2005), who lived within 50 km of a central monitoring station. We calculated the 1- to 7-day moving averages of fine particulate matter (PM2.5), black carbon (BC), sulfate (SO42-), nitrogen oxides (NOx), and ozone before examination visits. We used linear mixed effect models for P-selectin, monocyte chemoattractant protein 1 (MCP-1), intercellular adhesion molecule 1, lipoprotein-associated phospholipase A2 activity and mass, and osteoprotegerin that were measured up to twice, and linear regression models for CD40 ligand and interleukin-18 that were measured once, adjusting for demographics, life style and clinical factors, socioeconomic position, time, and meteorology. RESULTS We found negative associations of PM2.5 and BC with P-selectin, of ozone with MCP-1, and of SO42- and NOx with osteoprotegerin. At the 5-day moving average, a 5 µg/m3 higher PM2.5 was associated with 1.6% (95% CI: - 2.8, - 0.3) lower levels of P-selectin; a 10 ppb higher ozone was associated with 1.7% (95% CI: - 3.2, - 0.1) lower levels of MCP-1; and a 20 ppb higher NOx was associated with 2.0% (95% CI: - 3.6, - 0.4) lower levels of osteoprotegerin. CONCLUSIONS We did not find evidence of positive associations between short-term air pollution exposure and endothelial cell activation. On the contrary, short-term exposure to higher levels of ambient pollutants were associated with lower levels of P-selectin, MCP-1, and osteoprotegerin in the Framingham Heart Study.
Collapse
Affiliation(s)
- Wenyuan Li
- Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, United States; Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Kirsten S Dorans
- Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, United States; Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States; Tulane School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Elissa H Wilker
- Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, United States; Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Mary B Rice
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Petter L Ljungman
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States; Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Joel D Schwartz
- Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, United States
| | - Brent A Coull
- Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, United States
| | - Petros Koutrakis
- Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, United States
| | - Diane R Gold
- Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, United States; Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - John F Keaney
- University of Massachusetts Medical School, Worcester, MA, United States
| | - Ramachandran S Vasan
- Boston University Schools of Medicine and Public Health, Boston, MA, United States; National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, United States
| | - Emelia J Benjamin
- Boston University Schools of Medicine and Public Health, Boston, MA, United States; National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, United States
| | - Murray A Mittleman
- Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, United States; Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
16
|
Tibaut M, Caprnda M, Kubatka P, Sinkovič A, Valentova V, Filipova S, Gazdikova K, Gaspar L, Mozos I, Egom EE, Rodrigo L, Kruzliak P, Petrovic D. Markers of Atherosclerosis: Part 1 - Serological Markers. Heart Lung Circ 2018; 28:667-677. [PMID: 30468147 DOI: 10.1016/j.hlc.2018.06.1057] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/30/2018] [Accepted: 06/25/2018] [Indexed: 12/29/2022]
Abstract
Atherosclerosis is a major contributor to morbidity and mortality worldwide. With therapeutic consequences in mind, several risk scores are being used to differentiate individuals with low, intermediate or high cardiovascular (CV) event risk. The most appropriate management of intermediate risk individuals is still not known, therefore, novel biomarkers are being sought to help re-stratify them as low or high risk. This narrative review is presented in two parts. Here, in Part 1, we summarise current knowledge on serum (serological) biomarkers of atherosclerosis. Among novel biomarkers, high sensitivity C-reactive protein (hsCRP) has emerged as the most promising in chronic situations, others need further clinical studies. However, it seems that a combination of serum biomarkers offers more to risk stratification than either biomarker alone. In Part 2, we address genetic and imaging markers of atherosclerosis, as well as other developments relevant to risk prediction.
Collapse
Affiliation(s)
- Miha Tibaut
- General Hospital Murska Sobota, Murska Sobota, Slovenia
| | - Martin Caprnda
- 1st Department of Internal Medicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia; Department of Experimental Carcinogenesis, Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Andreja Sinkovič
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | | | - Slavomira Filipova
- Department of Cardiology, National Institute of Cardiovascular Diseases and Slovak Medical University, Bratislava, Slovakia
| | - Katarina Gazdikova
- Department of Nutrition, Faculty of Nursing and Professional Health Studies, Slovak Medical University, Bratislava, Slovakia; Department of General Medicine, Faculty of Medicine, Slovak Medical University, Bratislava, Slovakia.
| | - Ludovit Gaspar
- 1st Department of Internal Medicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia.
| | - Ioana Mozos
- Department of Functional Sciences, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania; Center for Translational Research and Systems Medicine, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Emmanuel E Egom
- Jewish General Hospital and Lady Davis Institute for Medical Research, Montreal, Canada; Department of Cardiology, The Adelaide and Meath Hospital Dublin, Incorporating the National Children Hospital, Dublin, Ireland
| | - Luis Rodrigo
- Faculty of Medicine, University of Oviedo, Central University Hospital of Asturias (HUCA), Oviedo, Spain
| | - Peter Kruzliak
- 2nd Department of Surgery, Center for Vascular Disease, Faculty of Medicine, Masaryk University and St. Anne's University Hospital, Brno, Czech Republic; Department of Internal Medicine, Brothers of Mercy Hospital, Brno, Czech Republic.
| | - Daniel Petrovic
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
17
|
Nativel M, Schneider F, Saulnier PJ, Gand E, Ragot S, Meilhac O, Rondeau P, Burillo E, Cournot M, Potier L, Velho G, Marre M, Roussel R, Rigalleau V, Mohammedi K, Hadjadj S. Prognostic Values of Inflammatory and Redox Status Biomarkers on the Risk of Major Lower-Extremity Artery Disease in Individuals With Type 2 Diabetes. Diabetes Care 2018; 41:2162-2169. [PMID: 30072406 DOI: 10.2337/dc18-0695] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/01/2018] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Inflammation and oxidative stress play an important role in the pathogenesis of lower-extremity artery disease (LEAD). We assessed the prognostic values of inflammatory and redox status biomarkers on the risk of LEAD in individuals with type 2 diabetes. RESEARCH DESIGN AND METHODS Plasma concentrations of tumor necrosis factor-α receptor 1 (TNFR1), angiopoietin-like 2, ischemia-modified albumin (IMA), fluorescent advanced glycation end products, protein carbonyls, and total reductive capacity of plasma were measured at baseline in the SURDIAGENE (Survie, Diabete de type 2 et Genetique) cohort. Major LEAD was defined as the occurrence during follow-up of peripheral revascularization or lower-limb amputation. RESULTS Among 1,412 participants at baseline (men 58.2%, mean [SD] age 64.7 [10.6] years), 112 (7.9%) developed major LEAD during 5.6 years of follow-up. High plasma concentrations of TNFR1 (hazard ratio [95% CI] for second vs. first tertile 1.12 [0.62-2.03; P = 0.71] and third vs. first tertile 2.16 [1.19-3.92; P = 0.01]) and of IMA (2.42 [1.38-4.23; P = 0.002] and 2.04 [1.17-3.57; P = 0.01], respectively) were independently associated with an increased risk of major LEAD. Plasma concentrations of TNFR1 but not IMA yielded incremental information, over traditional risk factors, for the risk of major LEAD as follows: C-statistic change (0.036 [95% CI 0.013-0.059]; P = 0.002), integrated discrimination improvement (0.012 [0.005-0.022]; P < 0.001), continuous net reclassification improvement (NRI) (0.583 [0.294-0.847]; P < 0.001), and categorical NRI (0.171 [0.027-0.317]; P = 0.02). CONCLUSIONS Independent associations exist between high plasma TNFR1 or IMA concentrations and increased 5.6-year risk of major LEAD in people with type 2 diabetes. TNFR1 allows incremental prognostic information, suggesting its use as a biomarker for LEAD.
Collapse
Affiliation(s)
- Mathilde Nativel
- Département d'Endocrinologie, Diabétologie, Nutrition, Hôpital Haut-Lévêque, Pessac, Bordeaux, France
| | - Fabrice Schneider
- Département de Chirurgie Vasculaire, CHU de Poitiers, Poitiers, France.,UFR de Médecine et Pharmacie, Université de Poitiers, Poitiers, France
| | - Pierre-Jean Saulnier
- UFR de Médecine et Pharmacie, Université de Poitiers, Poitiers, France.,Centre d'Investigation Clinique, CHU de Poitiers, Poitiers, France.,CIC 1402, INSERM, Poitiers, France
| | - Elise Gand
- Pôle Dune, CHU de Poitiers, Poitiers, France
| | - Stéphanie Ragot
- UFR de Médecine et Pharmacie, Université de Poitiers, Poitiers, France.,Centre d'Investigation Clinique, CHU de Poitiers, Poitiers, France.,CIC 1402, INSERM, Poitiers, France
| | - Olivier Meilhac
- UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, Université de La Réunion, Saint Denis de La Réunion, France.,CHU de La Réunion, Saint Denis de La Réunion, France
| | - Philippe Rondeau
- UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, Université de La Réunion, Saint Denis de La Réunion, France
| | - Elena Burillo
- UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, Université de La Réunion, Saint Denis de La Réunion, France
| | - Maxime Cournot
- UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, Université de La Réunion, Saint Denis de La Réunion, France.,Service de cardiologie, Centre Hospitalier Gabriel Martin, Saint-Paul, France
| | - Louis Potier
- DHU FIRE, Département d'Endocrinologie, Diabétologie, Nutrition, Assistance Publique Hôpitaux de Paris, Bichat Hospital, Paris, France.,UFR de Médecine, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,UMRS 1138, Centre de Recherche des Cordeliers, INSERM, Paris, France
| | - Gilberto Velho
- UMRS 1138, Centre de Recherche des Cordeliers, INSERM, Paris, France
| | - Michel Marre
- DHU FIRE, Département d'Endocrinologie, Diabétologie, Nutrition, Assistance Publique Hôpitaux de Paris, Bichat Hospital, Paris, France.,UFR de Médecine, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,UMRS 1138, Centre de Recherche des Cordeliers, INSERM, Paris, France
| | - Ronan Roussel
- DHU FIRE, Département d'Endocrinologie, Diabétologie, Nutrition, Assistance Publique Hôpitaux de Paris, Bichat Hospital, Paris, France.,UFR de Médecine, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,UMRS 1138, Centre de Recherche des Cordeliers, INSERM, Paris, France
| | - Vincent Rigalleau
- Département d'Endocrinologie, Diabétologie, Nutrition, Hôpital Haut-Lévêque, Pessac, Bordeaux, France.,Faculté de Médecine, Université de Bordeaux, Bordeaux, France.,Centre de Recherche INSERM-Université de Bordeaux U1219 "Bordeaux Population Health," Bordeaux, France
| | - Kamel Mohammedi
- Département d'Endocrinologie, Diabétologie, Nutrition, Hôpital Haut-Lévêque, Pessac, Bordeaux, France .,Faculté de Médecine, Université de Bordeaux, Bordeaux, France.,Centre de Recherche INSERM-Université de Bordeaux U1219 "Bordeaux Population Health," Bordeaux, France
| | - Samy Hadjadj
- UFR de Médecine et Pharmacie, Université de Poitiers, Poitiers, France.,CIC 1402, INSERM, Poitiers, France.,Département d'Endocrinologie, Diabétologie, Nutrition, CHU de Poitiers, Poitiers, France.,Research Unit 1082, INSERM, Poitiers, France
| |
Collapse
|
18
|
Nowobilski R, Kusinska K, Bukowska-Strakova K, Nizankowski R, Nowak W, Mika P, Jozkowicz A, Szczeklik A, Dulak J. Exercise training in intermittent claudication: Effects on antioxidant genes, inflammatory mediators and proangiogenic progenitor cells. Thromb Haemost 2017; 108:824-31. [DOI: 10.1160/th12-04-0278] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 07/28/2012] [Indexed: 01/13/2023]
Abstract
SummaryExercise training remains a therapy of choice in intermittent claudication (IC). However, too exhaustive exercise may cause ischaemic injury and inflammatory response. We tested the impact of three-month treadmill training and single treadmill exercise on antioxidant gene expressions, cytokine concentrations and number of marrow-derived proangiogenic progenitor cells (PPC) in the blood of IC patients. Blood samples of 12 patients were collected before and after training, before and 1, 3 and 6 hours after the single exercise. PPCs were analysed with flow cytometry, cytokine concentrations were checked with Milliplex MAP, while expression of mRNAs and miRNAs was evaluated with qRT-PCR. Treadmill training improved pain-free walking time (from 144 ± 44 seconds [s] to 311 ± 134 s, p=0.02) and maximum walking time (from 578 ± 293 s to 859 ± 423 s, p=0.01) in IC patients. Before, but not after training, the single treadmill exercise increased the number of circulating CD45dimCD34+CD133-KDR+ PPCs (p=0.048), decreased expression of HMOX1 (p=0.04) in circulating leukocytes, reduced tumour necrosis factor-α (p=0.03) and tended to elevate myeloperoxidase (p=0.06) concentrations in plasma. In contrast, total plasminogen activator inhibitor-1 was decreased by single exercise only after, but not before training (p=0.02). Both before and after training the single exercise decreased monocyte chemoattractant protein (MCP)-1 (p=0.006 and p=0.03) concentration and increased SOD1 (p=0.001 and p=0.01) expression. Patients after training had also less interleukin-6 (p=0.03), but more MCP-1 (p=0.04) in the blood. In conclusion, treadmill training improves walking performance of IC patients, attenuates the single exercise-induced changes in gene expressions or PPC mobilisation, but may also lead to higher production of some proinflammatory cytokines.
Collapse
|
19
|
Berardi C, Wassel CL, Decker PA, Larson NB, Kirsch PS, de Andrade M, Tsai MY, Pankow JS, Sale MM, Sicotte H, Tang W, Hanson NQ, McDermott MM, Criqui MH, Allison MA, Bielinski SJ. Elevated Levels of Adhesion Proteins Are Associated With Low Ankle-Brachial Index. Angiology 2017; 68:322-329. [PMID: 27436494 PMCID: PMC5247409 DOI: 10.1177/0003319716659178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Inflammation plays a pivotal role in peripheral artery disease (PAD). Cellular adhesion proteins mediate the interaction of leukocytes with endothelial cells during inflammation. To determine the association of cellular adhesion molecules with ankle-brachial index (ABI) and ABI category (≤1.0 vs >1.0) in a diverse population, 15 adhesion proteins were measured in the Multi-Ethnic Study of Atherosclerosis (MESA). To assess multivariable associations of each protein with ABI and ABI category, linear and logistic regression was used, respectively. Among 2364 participants, 23 presented with poorly compressible arteries (ABI > 1.4) and were excluded and 261 had ABI ≤ 1.0. Adjusting for traditional risk factors, elevated levels of soluble P-selectin, hepatocyte growth factor, and secretory leukocyte protease inhibitor were associated with lower ABI ( P = .0004, .001, and .002, respectively). Per each standard deviation of protein, we found 26%, 20%, and 19% greater odds of lower ABI category ( P = .001, .01, and .02, respectively). Further investigation into the adhesion pathway may shed new light on biological mechanisms implicated in PAD.
Collapse
Affiliation(s)
- Cecilia Berardi
- Department of Internal Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Christine L. Wassel
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Colchester, VT, USA
| | - Paul A. Decker
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | - Phillip S. Kirsch
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Mariza de Andrade
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Michael Y. Tsai
- Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - James S. Pankow
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA
| | - Michele M. Sale
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Hugues Sicotte
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Weihong Tang
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA
| | - Naomi Q. Hanson
- Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Mary M. McDermott
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Michael H. Criqui
- Department of Family Medicine and Public Health, University of California, San Diego, La Jolla, CA, USA
| | - Michael A. Allison
- Department of Family Medicine and Public Health, University of California, San Diego, La Jolla, CA, USA
| | | |
Collapse
|
20
|
Cauley JA, Kassem AM, Lane NE, Thorson S. Prevalent peripheral arterial disease and inflammatory burden. BMC Geriatr 2016; 16:213. [PMID: 27938334 PMCID: PMC5148838 DOI: 10.1186/s12877-016-0389-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 12/06/2016] [Indexed: 12/20/2022] Open
Abstract
Background Strong evidence implicates inflammation in the development of atherosclerotic heart disease but less is known about peripheral arterial disease (PAD). Our objective was to test the hypothesis that a composite index of inflammatory burden is associated with PAD. Methods Cross-sectional analysis of a randomly-selected group of 903 community-dwelling men in the MrOS cohort recruited between 2000 and 2002. Using blood samples, we measured seven cytokines and related these levels to prevalent PAD (ankle-brachial index (ABI) <0.9) both individually and as part of an “inflammatory burden score” (a composite sum of the number of pro-inflammatory cytokines in the highest quartile). Results Overall, 6.75% of men had ABI <0.9. The odds of prevalent PAD were higher in men with the highest quartile (Q4) levels of interleukin-6 multivariable (MV) adjusted (odds ratio (OR) =3.95 (95% CI, 1.4–11.3), tumor necrosis factor alpha OR = 4.44 (95% confidence interval (CI), 1.5–12.8), and C-reactive protein OR = 3.63 (95% CI, 1.4–9.4) compared to men in Q1. The magnitude of the association of these cytokines with PAD was similar to the effect of being 10 years older, OR = 2.41 (95% CI, 1.16–3.7). These significant effects persisted after additional MV adjustment for smoking except for CRP. Men with the highest inflammatory burden score (≥3) had 3.6 (95% CI, 1.5–8.7) increased odds of PAD, p trend = 0.03. After smoking adjustment the linear trend was borderline statistically significant (p trend = 0.10). Conclusion Inflammatory burden is associated with prevalent PAD, an association similar to aging 10 years. The inflammatory effects of smoking contributes to the underlying association between inflammation and PAD.
Collapse
Affiliation(s)
- Jane A Cauley
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, 130 DeSoto Street, A510 Crabtree Hall, Pittsburgh, PA, 15261, USA.
| | - Ahmed M Kassem
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, 130 DeSoto Street, A510 Crabtree Hall, Pittsburgh, PA, 15261, USA
| | - Nancy E Lane
- University of California Davis Health System, 4625 2nd Avenue, Suite 2006, Sacramento, CA, 95817, USA
| | - Sara Thorson
- Family Medicine of Southwest, Peacehealth Southwest Medical Center, 400 NE Mother Joseph Place, Vancouver, WA, 986664, USA
| | | |
Collapse
|
21
|
Sun Q, Zhou T, Niu J, Wu P, Guo Y, Yang Y. Comparison of protein-chip array analysis and traditional ELISAs for biomarker detection of diabetic limb arterial stenosis. Vascular 2016; 25:260-265. [PMID: 27771621 DOI: 10.1177/1708538116671078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The aim of this study was to screen the biomarkers of diabetic limb arterial stenosis. Fasting blood samples of 40 patients with diabetic limb arterial stenosis (experimental group), 40 diabetes patients (diabetic control group), and 40 healthy individuals (healthy control group) were collected. Protein-chip assay analysis and ELISA were used to detect tumor necrosis factor α, interleukin-6, endothelin-1, calcitonin gene-related peptide and high-sensitivity C-reactive protein in the three groups. Protein-chip array analysis and ELISA found consistent results that endothelin-1, tumor necrosis factor α, interleukin-6 and high-sensitivity C-reactive protein in the experimental group were significantly up-regulated while the expression of calcitonin gene-related peptide was down-regulated compared with the healthy control group ( P < 0.01). When compared with the diabetic control group, only markedly increased calcitonin gene-related peptide and interleukin-6 were observed in the experimental group ( P < 0.01). The study suggests that high-throughput protein-chip may be a reliable method to screen biomarkers of diabetic limb arterial stenosis. Calcitonin gene-related peptide and interleukin-6 might be promising biomarkers for diabetic limb arterial stenosis.
Collapse
Affiliation(s)
- Qiang Sun
- 1 Department of Peripheral Vascular, The Second Hospital of Shandong University, Jinan, Shandong Province, China
| | - Tao Zhou
- 1 Department of Peripheral Vascular, The Second Hospital of Shandong University, Jinan, Shandong Province, China
| | - Jun Niu
- 2 Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Peng Wu
- 1 Department of Peripheral Vascular, The Second Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yanan Guo
- 1 Department of Peripheral Vascular, The Second Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yanfei Yang
- 1 Department of Peripheral Vascular, The Second Hospital of Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
22
|
Garg PK, Arnold AM, Hinckley Stukovsky KD, Koro C, Jenny NS, Mukamal KJ, Criqui MH, Furberg CD, Newman AB, Cushman M. Lipoprotein-Associated Phospholipase A2 and Incident Peripheral Arterial Disease in Older Adults: The Cardiovascular Health Study. Arterioscler Thromb Vasc Biol 2016; 36:750-6. [PMID: 26848158 DOI: 10.1161/atvbaha.115.306647] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/25/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Although prior studies report a relationship between elevated lipoprotein-associated phospholipase A2 (Lp-PLA2) and incident cardiovascular disease, the prospective association of Lp-PLA2 with incident peripheral arterial disease (PAD) has not been studied. We investigated the association between Lp-PLA2 mass and activity and the risk of developing clinical PAD and low ankle-brachial index (ABI). APPROACH AND RESULTS Among Cardiovascular Health Study participants, a population-based cohort of 5888 adults aged ≥65 years enrolled in 1989 to 1990, Lp-PLA2 mass and activity were measured in 4537 individuals without baseline PAD. Clinical PAD, defined as leg artery revascularization or diagnosed claudication, was ascertained through 2011. Incident low ABI, defined as ABI <0.9 and decline of ≥0.15, was assessed among 3537 individuals who had an ABI >0.9 at baseline and a second ABI measurement 3 or 6 years later. Analyses were adjusted for demographics, cholesterol, smoking, comorbidities, and C-reactive protein. Each standard deviation increment in Lp-PLA2 mass (117 ng/mL) was associated with a higher risk of developing clinical PAD (hazard ratio 1.28; 95% confidence interval 1.13, 1.45) and incident low ABI (odds ratio 1.16; 95% confidence interval 1.00, 1.33). Results per standard deviation increment in Lp-PLA2 activity (13 nmol/min per mL) were similar for clinical PAD (hazard ratio 1.24; 95% confidence interval 1.07, 1.44) and low ABI (odds ratio 1.28; 95% confidence interval 1.09, 1.50). CONCLUSIONS Higher Lp-PLA2 mass and activity were associated with development of both incident clinical PAD and low ABI. Future studies are needed to determine whether pharmacological inhibition of Lp-PLA2 reduces the incidence of PAD.
Collapse
Affiliation(s)
- Parveen K Garg
- From the Division of Cardiology, University of Southern California Keck School of Medicine, Los Angeles (P.K.G.); Department of Biostatistics, University of Washington, Seattle (A.M.A., K.D.H.S.); Glaxo Smith Kline, Collegeville, PA (C.K.); Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington (N.S.J., M.C.); Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (K.J.M.); Department of Family and Preventive Medicine, University of California in San Diego School of Medicine, La Jolla (M.H.C.); Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC (C.D.F.); Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, PA (A.B.N.); and Department of Medicine, Cardiovascular Research Institute, University of Vermont College of Medicine, Burlington (M.C.).
| | - Alice M Arnold
- From the Division of Cardiology, University of Southern California Keck School of Medicine, Los Angeles (P.K.G.); Department of Biostatistics, University of Washington, Seattle (A.M.A., K.D.H.S.); Glaxo Smith Kline, Collegeville, PA (C.K.); Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington (N.S.J., M.C.); Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (K.J.M.); Department of Family and Preventive Medicine, University of California in San Diego School of Medicine, La Jolla (M.H.C.); Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC (C.D.F.); Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, PA (A.B.N.); and Department of Medicine, Cardiovascular Research Institute, University of Vermont College of Medicine, Burlington (M.C.)
| | - Karen D Hinckley Stukovsky
- From the Division of Cardiology, University of Southern California Keck School of Medicine, Los Angeles (P.K.G.); Department of Biostatistics, University of Washington, Seattle (A.M.A., K.D.H.S.); Glaxo Smith Kline, Collegeville, PA (C.K.); Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington (N.S.J., M.C.); Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (K.J.M.); Department of Family and Preventive Medicine, University of California in San Diego School of Medicine, La Jolla (M.H.C.); Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC (C.D.F.); Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, PA (A.B.N.); and Department of Medicine, Cardiovascular Research Institute, University of Vermont College of Medicine, Burlington (M.C.)
| | - Carol Koro
- From the Division of Cardiology, University of Southern California Keck School of Medicine, Los Angeles (P.K.G.); Department of Biostatistics, University of Washington, Seattle (A.M.A., K.D.H.S.); Glaxo Smith Kline, Collegeville, PA (C.K.); Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington (N.S.J., M.C.); Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (K.J.M.); Department of Family and Preventive Medicine, University of California in San Diego School of Medicine, La Jolla (M.H.C.); Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC (C.D.F.); Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, PA (A.B.N.); and Department of Medicine, Cardiovascular Research Institute, University of Vermont College of Medicine, Burlington (M.C.)
| | - Nancy S Jenny
- From the Division of Cardiology, University of Southern California Keck School of Medicine, Los Angeles (P.K.G.); Department of Biostatistics, University of Washington, Seattle (A.M.A., K.D.H.S.); Glaxo Smith Kline, Collegeville, PA (C.K.); Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington (N.S.J., M.C.); Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (K.J.M.); Department of Family and Preventive Medicine, University of California in San Diego School of Medicine, La Jolla (M.H.C.); Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC (C.D.F.); Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, PA (A.B.N.); and Department of Medicine, Cardiovascular Research Institute, University of Vermont College of Medicine, Burlington (M.C.)
| | - Kenneth J Mukamal
- From the Division of Cardiology, University of Southern California Keck School of Medicine, Los Angeles (P.K.G.); Department of Biostatistics, University of Washington, Seattle (A.M.A., K.D.H.S.); Glaxo Smith Kline, Collegeville, PA (C.K.); Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington (N.S.J., M.C.); Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (K.J.M.); Department of Family and Preventive Medicine, University of California in San Diego School of Medicine, La Jolla (M.H.C.); Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC (C.D.F.); Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, PA (A.B.N.); and Department of Medicine, Cardiovascular Research Institute, University of Vermont College of Medicine, Burlington (M.C.)
| | - Michael H Criqui
- From the Division of Cardiology, University of Southern California Keck School of Medicine, Los Angeles (P.K.G.); Department of Biostatistics, University of Washington, Seattle (A.M.A., K.D.H.S.); Glaxo Smith Kline, Collegeville, PA (C.K.); Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington (N.S.J., M.C.); Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (K.J.M.); Department of Family and Preventive Medicine, University of California in San Diego School of Medicine, La Jolla (M.H.C.); Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC (C.D.F.); Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, PA (A.B.N.); and Department of Medicine, Cardiovascular Research Institute, University of Vermont College of Medicine, Burlington (M.C.)
| | - Curt D Furberg
- From the Division of Cardiology, University of Southern California Keck School of Medicine, Los Angeles (P.K.G.); Department of Biostatistics, University of Washington, Seattle (A.M.A., K.D.H.S.); Glaxo Smith Kline, Collegeville, PA (C.K.); Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington (N.S.J., M.C.); Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (K.J.M.); Department of Family and Preventive Medicine, University of California in San Diego School of Medicine, La Jolla (M.H.C.); Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC (C.D.F.); Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, PA (A.B.N.); and Department of Medicine, Cardiovascular Research Institute, University of Vermont College of Medicine, Burlington (M.C.)
| | - Anne B Newman
- From the Division of Cardiology, University of Southern California Keck School of Medicine, Los Angeles (P.K.G.); Department of Biostatistics, University of Washington, Seattle (A.M.A., K.D.H.S.); Glaxo Smith Kline, Collegeville, PA (C.K.); Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington (N.S.J., M.C.); Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (K.J.M.); Department of Family and Preventive Medicine, University of California in San Diego School of Medicine, La Jolla (M.H.C.); Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC (C.D.F.); Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, PA (A.B.N.); and Department of Medicine, Cardiovascular Research Institute, University of Vermont College of Medicine, Burlington (M.C.)
| | - Mary Cushman
- From the Division of Cardiology, University of Southern California Keck School of Medicine, Los Angeles (P.K.G.); Department of Biostatistics, University of Washington, Seattle (A.M.A., K.D.H.S.); Glaxo Smith Kline, Collegeville, PA (C.K.); Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington (N.S.J., M.C.); Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (K.J.M.); Department of Family and Preventive Medicine, University of California in San Diego School of Medicine, La Jolla (M.H.C.); Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC (C.D.F.); Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, PA (A.B.N.); and Department of Medicine, Cardiovascular Research Institute, University of Vermont College of Medicine, Burlington (M.C.)
| |
Collapse
|
23
|
Engelberger RP, Limacher A, Kucher N, Baumann F, Silbernagel G, Benghozi R, Do DD, Willenberg T, Baumgartner I. Biological variation of established and novel biomarkers for atherosclerosis: Results from a prospective, parallel-group cohort study. Clin Chim Acta 2015; 447:16-22. [DOI: 10.1016/j.cca.2015.05.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 05/05/2015] [Accepted: 05/06/2015] [Indexed: 01/26/2023]
|
24
|
A review of the pathophysiology and potential biomarkers for peripheral artery disease. Int J Mol Sci 2015; 16:11294-322. [PMID: 25993296 PMCID: PMC4463701 DOI: 10.3390/ijms160511294] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 03/29/2015] [Accepted: 04/08/2015] [Indexed: 12/12/2022] Open
Abstract
Peripheral artery disease (PAD) is due to the blockage of the arteries supplying blood to the lower limbs usually secondary to atherosclerosis. The most severe clinical manifestation of PAD is critical limb ischemia (CLI), which is associated with a risk of limb loss and mortality due to cardiovascular events. Currently CLI is mainly treated by surgical or endovascular revascularization, with few other treatments in routine clinical practice. There are a number of problems with current PAD management strategies, such as the difficulty in selecting the appropriate treatments for individual patients. Many patients undergo repeated attempts at revascularization surgery, but ultimately require an amputation. There is great interest in developing new methods to identify patients who are unlikely to benefit from revascularization and to improve management of patients unsuitable for surgery. Circulating biomarkers that predict the progression of PAD and the response to therapies could assist in the management of patients. This review provides an overview of the pathophysiology of PAD and examines the association between circulating biomarkers and PAD presence, severity and prognosis. While some currently identified circulating markers show promise, further larger studies focused on the clinical value of the biomarkers over existing risk predictors are needed.
Collapse
|
25
|
Wassel CL, Berardi C, Pankow JS, Larson NB, Decker PA, Hanson NQ, Tsai MY, Criqui MH, Allison MA, Bielinski SJ. Soluble P-selectin predicts lower extremity peripheral artery disease incidence and change in the ankle brachial index: the Multi-Ethnic Study of Atherosclerosis (MESA). Atherosclerosis 2015; 239:405-11. [PMID: 25682040 PMCID: PMC4361295 DOI: 10.1016/j.atherosclerosis.2015.01.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 01/04/2015] [Accepted: 01/19/2015] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To determine the association of circulating P-selectin with prevalent and incident peripheral artery disease (PAD), the ankle brachial index (ABI), and change in the ABI. METHODS The Multi-Ethnic Study of Atherosclerosis (MESA) is a prospective population-based cohort study including 6814 European descent, African American, Hispanic and Chinese men and women aged 45-84 at baseline. Four clinical exams took place after the baseline exam. After excluding those with ABI>1.4, prevalent and incident PAD were defined as an ABI≤0.90. ABI progression was defined as progression from a normal ABI (0.91-1.4) to abnormal (≤0.90 or >1.4) at a later exam. RESULTS In adjusted models, each SD (13 ng/mL) higher P-selectin was significantly associated with 0.007 lower ABI (95% CI ((-0.011, -0.004)), p < 0.001), and an average change in the ABI of -0.006 ((-0.010, -0.003, p < 0.001). P-selectin was significantly associated with a 1.17-fold greater odds of prevalent PAD ((1.02, 1.33), p = 0.03), and a 30% greater risk of incident PAD ((1.11, 1.53), p = 0.001), as well as progression from a normal ABI to an ABI≤ 0.90 (p = 0.003), but not to an ABI>1.4 (p = 0.96). Addition of P-selectin to models containing traditional PAD risk factors and markers of inflammation/coagulation significantly improved the net reclassification for ABI progression (p = 0.03), but was only marginally significant for incident PAD (p = 0.06). CONCLUSIONS P-selectin is significantly associated with the development of PAD. However, further research is needed in population-based studies to confirm prospective associations of P-selectin with incident PAD and change in the ABI, as well as its potential predictive ability.
Collapse
Affiliation(s)
- Christina L. Wassel
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - Cecilia Berardi
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Nicholas B. Larson
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Paul A. Decker
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Naomi Q. Hanson
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, MN
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, MN
| | - Michael H. Criqui
- Division of Preventive Medicine, Department of Family and Preventive Medicine, University of California – San Diego, La Jolla, CA
| | - Matthew A. Allison
- Division of Preventive Medicine, Department of Family and Preventive Medicine, University of California – San Diego, La Jolla, CA
| | - Suzette J. Bielinski
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| |
Collapse
|
26
|
Racial Differences in the Ability of Subclinical Atherosclerosis Testing to Predict CVD. CURRENT CARDIOVASCULAR RISK REPORTS 2015. [DOI: 10.1007/s12170-015-0453-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
27
|
Nakamura Y, Kunii H, Yoshihisa A, Takiguchi M, Shimizu T, Yamauchi H, Iwaya S, Owada T, Abe S, Sato T, Suzuki S, Oikawa M, Kobayashi A, Yamaki T, Sugimoto K, Nakazato K, Suzuki H, Saitoh SI, Takeishi Y. Impact of peripheral artery disease on prognosis in hospitalized heart failure patients. Circ J 2015; 79:785-93. [PMID: 25739573 DOI: 10.1253/circj.cj-14-1280] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND The impact of peripheral artery disease (PAD) on heart failure (HF) prognosis remains unclear. METHODS AND RESULTS A total of 388 consecutive decompensated HF patients were divided into 2 groups based on the presence of PAD: HF with PAD (PAD group, n=101, 26.0%) and HF without PAD (non-PAD group, n=287, 74.0%). We compared clinical features, echocardiographic parameters, cardiopulmonary exercise testing results, laboratory findings, as well as cardiac, non-cardiac, and all-cause mortality between the 2 groups. The PAD group, as compared with the non-PAD group, had (1) higher prevalence of coronary artery disease (40.6 vs. 27.5%, P=0.011) and cerebrovascular disease (34.7 vs. 18.2%, P=0.001); (2) higher tumor necrosis factor-α (1.82 vs. 1.49 pg/ml, P=0.023), C-reactive protein (0.32 vs. 0.19 mg/dl, P=0.045), and troponin T (0.039 vs. 0.021 ng/ml, P=0.019); (3) lower LVEF (42.4 vs. 48.5%, P<0.001); (4) lower peak V̇O2(13.4 vs. 15.9 ml·kg(-1)·min(-1), P=0.001); and (5) higher V̇E/V̇CO2slope (38.8 vs. 33.7, P<0.001). On Kaplan-Meier analysis, cardiac, non-cardiac, and all-cause mortality were significantly higher in the PAD group than in the non-PAD group (P<0.05, respectively). On Cox proportional hazard analysis after adjusting for confounding factors, PAD was an independent predictor of cardiac and all-cause mortality (P<0.05, respectively) in HF patients. CONCLUSIONS PAD was common and an independent predictor of cardiac and all-cause mortality in HF patients.
Collapse
Affiliation(s)
- Yuichi Nakamura
- Department of Cardiology and Hematology, Fukushima Medical University
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Rull A, Hernandez-Aguilera A, Fibla M, Sepulveda J, Rodríguez-Gallego E, Riera-Borrull M, Sirvent JJ, Martín-Paredero V, Menendez JA, Camps J, Joven J. Understanding the role of circulating chemokine (C-C motif) ligand 2 in patients with chronic ischemia threatening the lower extremities. Vasc Med 2014; 19:442-51. [PMID: 25336430 DOI: 10.1177/1358863x14554034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The role of chemokine (C-C motif) ligand 2 (CCL2) in peripheral artery disease is unclear. We measured the difference between serum and plasma levels of CCL2 in patients with chronic ischemia threatening the lower extremities following the observation that atypical chemokine receptors in blood and tissue cells may prevent CCL2 from entering the circulation and consequently modulate its function of attracting monocytes to the site of lesion. To identify the influence of CCL2, we compared the patients' values to those in bio-banked samples from a control population. Further, we explored the association with the Asp42Gly polymorphism (rs12075) in Duffy antigen chemokine receptor; one of these atypical chemokine receptors. When possible, we evaluated in surgically excised normal and affected arteries the calcium burden as well as the expression of CCL2 and related receptors reflecting the inflammatory status. Our findings indicate that circulating CCL2 was significantly associated with the severity and presence of the disease (OR 0.966, 95% CI 0.944 to 0.988, p = 0.003). Circulating CCL2 was dependent on the rs12075 genotype (AA>AG>GG), which, probably, indicates a higher expression of chemokine receptor in the arteries of AA subjects. The associations with genetic variants and the over-expression of atypical chemokine receptors in diseased arteries may have potential implications and our data indicate that CCL2 may represent a previously unrecognized factor that needs to be considered in the screening of patients with risk factors for peripheral artery disease.
Collapse
Affiliation(s)
- Anna Rull
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Anna Hernandez-Aguilera
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain Servei d'Angiologia, Cirurgia Vascular i Endovascular, Hospital Universitari Joan XXIII, Tarragona, Spain
| | - Montserrat Fibla
- Servei d'Anatomia Patològica, Hospital Universitari Joan XXIII, Tarragona, Spain
| | - Julio Sepulveda
- Servei d'Angiologia, Cirurgia Vascular i Endovascular, Hospital Universitari Joan XXIII, Tarragona, Spain
| | - Esther Rodríguez-Gallego
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Marta Riera-Borrull
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Juan J Sirvent
- Servei d'Anatomia Patològica, Hospital Universitari Joan XXIII, Tarragona, Spain
| | - Vicente Martín-Paredero
- Servei d'Angiologia, Cirurgia Vascular i Endovascular, Hospital Universitari Joan XXIII, Tarragona, Spain
| | - Javier A Menendez
- Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| |
Collapse
|
29
|
Callegari A, Coons ML, Ricks JL, Rosenfeld ME, Scatena M. Increased calcification in osteoprotegerin-deficient smooth muscle cells: Dependence on receptor activator of NF-κB ligand and interleukin 6. J Vasc Res 2014; 51:118-31. [PMID: 24642764 DOI: 10.1159/000358920] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 01/14/2014] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Vascular calcification is highly correlated with cardiovascular disease morbidity and mortality. Osteoprotegerin (OPG) is a secreted decoy receptor for receptor activator of NF-κB ligand (RANKL). Inactivation of OPG in apolipoprotein E-deficient (ApoE-/-) mice increases lesion size and calcification. The mechanism(s) by which OPG is atheroprotective and anticalcific have not been entirely determined. We investigated whether OPG-deficient vascular smooth muscle cells (VSMCs) are more susceptible to mineralization and whether RANKL mediates this process. RESULTS Lesion-free aortas from 12-week-old ApoE-/-OPG-/- mice had spotty calcification, an appearance of osteochondrogenic factors and a decrease of smooth muscle markers when compared to ApoE-/-OPG+/+ aortas. In osteogenic conditions, VSMCs isolated from ApoE-/-OPG-/- (KO-VSMC) mice deposited more calcium than VSMCs isolated from ApoE-/-OPG+/+ (WT-VSMC) mice. Gene expression and biochemical analysis indicated accelerated osteochondrogenic differentiation. Ablation of RANKL signaling in KO-VSMCs rescued the accelerated calcification. While WT-VSMCs did not respond to RANKL treatment, KO-VSMCs responded with enhanced calcification and the upregulation of osteochondrogenic genes. RANKL strongly induced interleukin 6 (IL-6), which partially mediated RANKL-dependent calcification and gene expression in KO-VSMCs. CONCLUSIONS OPG inhibits vascular calcification by regulating the procalcific effects of RANKL on VSMCs and is thus a possible target for therapeutic intervention.
Collapse
Affiliation(s)
- Andrea Callegari
- Department of Bioengineering, University of Washington, Seattle, Wash., USA
| | | | | | | | | |
Collapse
|
30
|
Hong SN, Gona P, Fontes JD, Oyama N, Chan RH, Kenchaiah S, Tsao CW, Yeon SB, Schnabel RB, Keaney JF, O'Donnell CJ, Benjamin EJ, Manning WJ. Atherosclerotic biomarkers and aortic atherosclerosis by cardiovascular magnetic resonance imaging in the Framingham Heart Study. J Am Heart Assoc 2013; 2:e000307. [PMID: 24242683 PMCID: PMC3886740 DOI: 10.1161/jaha.113.000307] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The relations between subclinical atherosclerosis and inflammatory biomarkers have generated intense interest but their significance remains unclear. We sought to determine the association between a panel of biomarkers and subclinical aortic atherosclerosis in a community-based cohort. METHODS AND RESULTS We evaluated 1547 participants of the Framingham Heart Study Offspring cohort who attended the 7th examination cycle and underwent both cardiovascular magnetic resonance imaging (CMR) and assays for 10 biomarkers associated with atherosclerosis: high-sensitivity C-reactive protein, fibrinogen, intercellular adhesion molecule-1, interleukin-6, interleukin-18, lipoprotein-associated phospholipase-A2 activity and mass, monocyte chemoattractant protein-1, P-selectin, and tumor necrosis factor receptor-2. In logistic regression analysis, we found no significant association between the biomarker panel and the presence of aortic plaque (global P=0.53). Using Tobit regression with aortic plaque as a continuous variable, we noted a modest association between biomarker panel and aortic plaque volume in age- and sex-adjusted analyses (P=0.003). However, this association was attenuated after further adjustment for clinical covariates (P=0.09). CONCLUSIONS In our community-based cohort, we found no significant association between our multibiomarker panel and aortic plaque. Our results underscore the strengths and limitations of the use of biomarkers for the identification of subclinical atherosclerosis and the importance of traditional risk factors.
Collapse
Affiliation(s)
- Susie N Hong
- Cardiovascular Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Dang Y, Xia Y, Li Y, Yu DCW. Anemia and type 2 diabetes mellitus associated with peripheral arterial disease progression in Chinese male patients. Clin Biochem 2013; 46:1673-7. [PMID: 23892184 DOI: 10.1016/j.clinbiochem.2013.07.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 07/10/2013] [Accepted: 07/12/2013] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Peripheral arterial disease (PAD) is a chronic occlusive disease mainly occurred in elderly adults. Arteriosclerosis obliterans (ASO) mainly occurring from small or medium sized arteries of the lower extremities is one of the most common causes of PAD. The gender-related differences of circulating risk factors in diabetic patients with ASO in China remain unknown. The aim of this study is to investigate the gender-related differences in the pattern of several potential risk factors between male and female patients with ASO and type 2 diabetes mellitus (T2D). DESIGN AND METHODS Clinical profiles and risk factors were analyzed in 323 Chinese patients with ASO and 112 patients were confirmed with T2D. Severities of limb ischemia were staged according to Fontaine classification. RESULTS The significant inverse correlation was seen between the increased age and hemoglobin. The significant positive correlation was seen between the increased age, urea and creatinine both in the non-diabetic and diabetic male patients. The expression levels of hemoglobin significantly correlate with the classification of Fontaine clinical symptoms in Chinese male patients with T2D/ASO. CONCLUSION The study is the first report indicating that the gender-related differences of circulating risk factors are associated with T2D patients with ASO in China. Anemia in Chinese male patients with T2D/ASO may play an important role in peripheral arterial disease progression.
Collapse
Affiliation(s)
- Yiping Dang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | |
Collapse
|
32
|
Schnabel RB, Yin X, Larson MG, Yamamoto JF, Fontes JD, Kathiresan S, Rong J, Levy D, Keaney JF, Wang TJ, Murabito JM, Vasan RS, Benjamin EJ. Multiple inflammatory biomarkers in relation to cardiovascular events and mortality in the community. Arterioscler Thromb Vasc Biol 2013; 33:1728-33. [PMID: 23640499 DOI: 10.1161/atvbaha.112.301174] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Evidence suggests that chronic low-grade inflammation and oxidative stress are related to cardiovascular disease (CVD) and mortality. APPROACH AND RESULTS We examined 11 established and novel biomarkers representing inflammation and oxidative stress (C-reactive protein, fibrinogen, interleukin-6, intercellular adhesion molecule-1, lipoprotein-associated phospholipase-A2 [mass and activity], monocyte chemoattractant protein-1, myeloperoxidase, CD40 ligand, P-selectin, and tumor necrosis factor receptor II [TNFRII]) in relation to incident major CVD and mortality in the community. We studied 3035 participants (mean age, 61 ± 9 years; 53% women). During follow-up (median, 8.9 years), 253 participants experienced a CVD event and 343 died. C-reactive protein (hazard ratio [HR] reported per SD ln-transformed biomarker, 1.18; 95% confidence interval [CI], 1.02-1.35; nominal P=0.02) and TNFRII (HR, 1.15; 95% CI, 1.01-1.32; nominal P=0.04) were retained in multivariable-adjusted models for major CVD, but were not significant after adjustment for multiple testing. The biomarkers related to mortality were TNFRII (HR, 1.33; 95% CI, 1.19-1.49; P<0.0001), ICAM-1 (HR, 1.24; 95% CI, 1.12-1.37; P<0.0001), and interleukin-6 (HR, 1.25; 95% CI, 1.12-1.39; P<0.0001). The addition of these markers to the model, including traditional risk factors, increased discrimination and reclassification for risk of death (P<0.0001), but not for CVD. CONCLUSIONS Of 11 inflammatory biomarkers tumor necrosis factor receptor II was related to cardiovascular disease and mortality in the Framingham Heart Study. The combination of TNFRII with C-reactive protein in relation to CVD and with interleukin-6 to mortality increased the predictive ability in addition to CVD risk factors for total mortality but not for incident CVD.
Collapse
Affiliation(s)
- Renate B Schnabel
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Fontes JD, Yamamoto JF, Larson MG, Wang N, Dallmeier D, Rienstra M, Schnabel RB, Vasan RS, Keaney JF, Benjamin EJ. Clinical correlates of change in inflammatory biomarkers: The Framingham Heart Study. Atherosclerosis 2013; 228:217-23. [PMID: 23489346 PMCID: PMC3650714 DOI: 10.1016/j.atherosclerosis.2013.01.019] [Citation(s) in RCA: 42] [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: 04/17/2012] [Revised: 01/05/2013] [Accepted: 01/16/2013] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Traditional clinical risk factors are associated with inflammation cross-sectionally, but associations of longitudinal variation in inflammatory biomarkers with corresponding changes in clinical risk factors are incompletely described. We sought to analyze clinical factors associated with change in inflammation in the community. METHODS We studied 3013 Framingham Offspring (n = 2735) and Omni Cohort (n = 278) participants (mean age 59 years, 55% women, 9% ethnic/racial minority) who attended two consecutive examination cycles (mean 6.7 years apart). We selected ten inflammatory biomarkers representing distinctive biological functions: C-reactive protein (CRP), intercellular adhesion molecule-1, interleukin-6, isoprostanes, lipoprotein-associated phospholipase-2 (Lp-PLA2) activity, Lp-PLA2-mass, monocyte chemoattractant protein-1, osteoprotegerin, P-selectin, and tumor necrosis factor receptor II (TNFRII). We constructed multivariable-adjusted regression models to assess the relations of baseline, follow-up and change in clinical risk factors with change in biomarker concentrations over time. RESULTS Baseline, follow-up and change in clinical risk factors explain a moderate amount of the variation in biomarker concentrations across 2 consecutive examinations (ranging from r(2) = 0.28 [TNFRII] up to 0.52 [Lp-PLA2-mass]). In multivariable models, increasing body-mass index, smoking initiation, worsening lipid profile, and increasing waist size were associated with increasing concentrations of several biomarkers. Conversely, hypercholesterolemia therapy and hormone replacement cessation were associated with decreasing concentrations of biomarkers such as CRP, Lp-PLA2-mass and activity. CONCLUSION Cardiovascular risk factors have different patterns of association with longitudinal change in inflammatory biomarkers and explain modest amounts of variability in biomarker concentrations. Nevertheless, a substantial proportion of longitudinal change in inflammatory markers is not explained by traditional risk factors.
Collapse
Affiliation(s)
- Joao D. Fontes
- Framingham Heart Study, Framingham, MA, USA
- Cardiology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | | | - Martin G. Larson
- Framingham Heart Study, Framingham, MA, USA
- Department of Mathematics and Statistics, Boston University, Boston, MA, USA
| | - Na Wang
- Department of Mathematics and Statistics, Boston University, Boston, MA, USA
| | - Dhayana Dallmeier
- Framingham Heart Study, Framingham, MA, USA
- Division of General Internal Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Michiel Rienstra
- Framingham Heart Study, Framingham, MA, USA
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Renate B. Schnabel
- Framingham Heart Study, Framingham, MA, USA
- Department of Medicine II, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Ramachandran S. Vasan
- Framingham Heart Study, Framingham, MA, USA
- Cardiology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
- Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - John F. Keaney
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Emelia J. Benjamin
- Framingham Heart Study, Framingham, MA, USA
- Cardiology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
- Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
34
|
Ye Z, Ali Z, Klee GG, Mosley TH, Kullo IJ. Associations of candidate biomarkers of vascular disease with the ankle-brachial index and peripheral arterial disease. Am J Hypertens 2013; 26:495-502. [PMID: 23467205 DOI: 10.1093/ajh/hps073] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The use of multiple biomarkers representing various etiologic pathways of atherosclerosis may improve the prediction of interindividual variation in the ankle-brachial index (ABI). To this end, we investigated associations of 47 candidate biomarkers with the ABI and presence of peripheral arterial disease (PAD) in African-Americans (AAs) and non-Hispanic whites (NHWs). METHODS Study participants included 1,291 AAs (71.1% women, mean age, 63.4±9.3 years) and 1,152 NHWs (57.5% women, mean age 58.5±10.1 years) belonging to hypertensive sibships. Peripheral arterial disease was defined as an ABI ≤ 0.90. Circulating levels of 47 candidate biomarkers were log-transformed before analysis because of skewed distribution. Multivariate regression analyses were used to identify biomarkers associated with ABI or PAD independently of age, sex, conventional risk factors, and medication use. RESULTS After adjustment for covariates, higher levels of nine biomarkers were associated with a lower ABI in AAs (all P ≤ 0.005); these biomarkers were C-reactive protein (CRP), interleukin-6, tumor necrosis factor receptor-II (TNF-R II), lipoprotein(a), N-terminal pro-brain natriuretic peptide (NT-proBNP), pro-atrial natriuretic peptide, C-terminal pro-arginine vasopressin, osteoprotegerin, and fibrinogen. Three biomarkers - myeloperoxidase, NT-proBNP, and D-dimer - were associated with ABI in NHWs (all P ≤ 0.01). C-reactive protein, interleukin-6, TNF-R II, lipoprotein(a), NT-proBNP, pro-atrial natriuretic peptide, D-dimer, and fibrinogen were associated with PAD (all P ≤ 0.005) in AAs after adjustment for covariates. None of the biomarkers were independently associated with PAD in NHWs. CONCLUSION A multimarker approach improved the prediction of interindividual variation in the ABI in AAs and NHWs, and improved prediction of the presence of PAD in AAs.
Collapse
Affiliation(s)
- Zi Ye
- Division of Cardiovascular Diseases and the Gonda Vascular Center, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | |
Collapse
|
35
|
Monopoli DE, Bertelli L, Sgura FA, Politi L, Becirovic M, Iaccarino D, Lattanzi A, Rampino K, Gorlato G, Menozzi M, Zennaro RG, Rossi R. Long term prognostic value of subclinical carotid and femoral arterial wall lesions in patients with ST-elevation-myocardial infarction having percutaneous coronary intervention. Am J Cardiol 2013; 111:649-56. [PMID: 23246270 DOI: 10.1016/j.amjcard.2012.11.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2012] [Revised: 11/12/2012] [Accepted: 11/12/2012] [Indexed: 12/15/2022]
Abstract
The presence of clinical peripheral arterial disease (PAD) is associated with an increased risk for adverse cardiovascular outcomes in patients with coronary artery disease. However, there are few data regarding the impact of the presence and degree of the subclinical PAD on outcomes in patients with coronary artery disease. The aim of this study was to assess prospectively the grade of subclinical PAD in the setting of patients who underwent primary percutaneous coronary intervention for the prediction of intermediate- and long-term clinical outcomes. A total of 971 consecutive patients without histories of clinical PAD who under went primary percutaneous coronary intervention for ST-segment elevation myocardial infarction were included in a prospective follow-up. Subclinical PAD severity was blindly assessed on the basis of an ultrasound arterial morphologic classification defined with the assessment of wall carotid and femoral artery bifurcations. This classification included 4 increasing classes of subclinical carotid and femoral arterial wall lesions, and the total group was divided accordingly. Death and major cardiovascular and cerebrovascular events were evaluated. During a median follow-up period of 40 months, a total of 109 patients (11.2%) died, 9 (2.8%) in class I, 12 (3.1%) in class II, 37 (23.7%) in class III, and 51 (49.0%) in class IV (p <0.001). On multivariate analysis, mortality in class IV was sevenfold higher (hazard ratio [HR] 7.34, 95% confidence interval [CI] 3.3 to 16.33, p <0.001) compared to class I and was also increased in class III (HR 5.38, 95% CI 2.42 to 11.92, p <0.001). Similar results were obtained for major adverse cardiovascular and cerebrovascular events in class IV (HR 7.50, 95% confidence interval 5.36 to 10.50, p <0.0001), class III (HR 6.44, 95% CI 4.45 to 9.32, p <0.001), and class II (HR 1.73, 95% CI 1.23 to 2.43, p = 0.002). In conclusion, ultrasound arterial morphologic classification may be applied in patients with ST-segment elevation myocardial infarctions who undergo primary percutaneous coronary intervention and can stratify patients for poor clinical outcomes during long-term follow-up.
Collapse
|
36
|
Singh S, Bailey KR, Kullo IJ. Ethnic differences in ankle brachial index are present in middle-aged individuals without peripheral arterial disease. Int J Cardiol 2013; 162:228-33. [PMID: 21652099 PMCID: PMC3174274 DOI: 10.1016/j.ijcard.2011.05.068] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2010] [Revised: 05/02/2011] [Accepted: 05/13/2011] [Indexed: 12/14/2022]
Abstract
INTRODUCTION To better understand the basis for previously reported ethnic differences in ankle brachial index (ABI), we investigated whether these differences were present in individuals without known peripheral arterial disease (PAD). METHODS We used data from National Health and Nutrition Examination surveys (NHANES 1999-2004) to determine whether ethnic differences were present in respondents without PAD (1 ≤ ABI ≤ 1.3). We assessed whether ethnicity was an independent predictor of ABI and ankle systolic blood pressure (SBP) in linear regression models that adjusted for conventional and novel cardiovascular risk factors. To minimize effects of atherosclerosis on ABI, we studied adults aged ≤ 60 years, and also repeated our analyses in a subset aged ≤ 50 years that did not have risk factors for PAD. RESULTS 3348 participants aged ≤ 60 years were included in the study. Mean ABI was 1.11 in non-Hispanic Blacks (NHB) and 1.13 in non-Hispanic Whites (NHW) (P < 0.0001). In multivariable linear regression analysis that adjusted for age, gender, ethnicity, smoking, height, diabetes, brachial SBP, dyslipidemia, diabetes, renal function, concurrent cardiovascular disease, and plasma levels of homocysteine, fibrinogen and C-reactive protein, NHB had lower ABI than NHW (β = -0.03 ± 0.004, P < 0.00001). Although, NHBs had higher ankle SBP than NHWs (by 5.4 mm Hg), NHBs had a lower mean ankle SBP (β = -3.663 mm Hg ± 0.500, P < 0.0001) after adjusting for clinical covariates, including brachial SBP, in multivariable analysis. CONCLUSION Ethnic differences in ABI are present in middle-aged adults at low risk for peripheral atherosclerosis.
Collapse
Affiliation(s)
- Siddharth Singh
- Division of Cardiovascular Diseases, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Kent R. Bailey
- Division of Biostatistics and Informatics, Health Services Research, Mayo Clinic, Rochester
| | | |
Collapse
|
37
|
Foster MC, Ghuman N, Hwang SJ, Murabito JM, Fox CS. Low ankle-brachial index and the development of rapid estimated GFR decline and CKD. Am J Kidney Dis 2012; 61:204-10. [PMID: 22901770 DOI: 10.1053/j.ajkd.2012.07.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 07/11/2012] [Indexed: 02/04/2023]
Abstract
BACKGROUND Low ankle-brachial index (ABI) is associated with increases in serum creatinine level. Whether low ABI is associated with the development of rapid estimated glomerular filtration rate (eGFR) decline, stage 3 chronic kidney disease (CKD), or microalbuminuria is uncertain. STUDY DESIGN Prospective cohort study. SETTING & PARTICIPANTS Framingham Offspring cohort participants who attended the sixth (1995-1998) and eighth (2005-2008) examinations. PREDICTOR ABI, categorized as normal (>1.1 to <1.4), low-normal (>0.9 to 1.1), and low (≤0.9). OUTCOMES Rapid eGFR decline (eGFR decline ≥3 mL/min/1.73 m(2) per year), incident stage 3 CKD (eGFR <60 mL/min/1.73 m(2)), incident microalbuminuria. MEASUREMENTS GFR was estimated using the serum creatinine-based CKD-EPI (CKD Epidemiology Collaboration) equation. Urinary albumin-creatinine ratio (UACR) was determined based on spot urine samples. RESULTS During 9.5 years, 9.0% (232 of 2,592) experienced rapid eGFR decline and 11.1% (270 of 2,426) developed stage 3 CKD. Compared to normal ABI, low ABI was associated with 5.73-fold increased odds of rapid eGFR decline (95% CI, 2.77-11.85; P<0.001) after age, sex, and baseline eGFR adjustment; this persisted after multivariable adjustment for standard CKD risk factors (OR, 3.60; 95% CI, 1.65-7.87; P = 0.001). After adjustment for age, sex, and baseline eGFR, low ABI was associated with a 2.51-fold increased odds of stage 3 CKD (OR, 2.51; 95% CI, 1.16-5.44; P = 0.02), although this was attenuated after multivariable adjustment (OR, 1.68; 95% CI, 0.75-3.76; P = 0.2). In 1,902 free of baseline microalbuminuria, low ABI was associated with increased odds of microalbuminuria after adjustment for age, sex, and baseline UACR (OR, 2.81; 95% CI, 1.07-7.37; P = 0.04), with attenuation upon further adjustment (OR, 1.88; P = 0.1). LIMITATIONS Limited number of events with low ABI. Outcomes based on single serum creatinine and UACR measurements at each examination. CONCLUSIONS Low ABI is associated with an increased risk of rapid eGFR decline, suggesting that systemic atherosclerosis predicts a decrease in kidney function.
Collapse
Affiliation(s)
- Meredith C Foster
- National Heart, Lung, and Blood Institute's (NHLBI's) Framingham Heart Study, Framingham, MA, USA
| | | | | | | | | |
Collapse
|
38
|
Patients with unrecognized peripheral arterial disease (PAD) assessed by ankle-brachial index (ABI) present a defined profile of proinflammatory markers compared to healthy subjects. Cytokine 2012; 59:294-8. [PMID: 22595645 DOI: 10.1016/j.cyto.2012.04.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 04/19/2012] [Accepted: 04/25/2012] [Indexed: 12/18/2022]
Abstract
BACKGROUND Many studies have postulated that atherosclerosis should be considered as an inflammatory disease. In addition, some studies have focused on the relationship between inflammation and peripheral arterial disease (PAD). OBJECTIVE Define the plasma levels of soluble markers, including the proinflammatory cytokine interleukin-6 (IL-6), the anti-inflammatory cytokine transforming growth factor-β1 (TGF-β1), the endothelial-specific adhesion factor (E-selectin) and two proteinases involved in extracellular matrix degradation (matrix metalloproteinases-2 and -9, MMP-2, and MMP-9) in previously unrecognized patients with peripheral artery disease (PAD) and non-PAD controls. RESULTS Significantly higher levels of IL-6, E-selectin and MMP-2/MMP-9 and significantly reduced levels of TGF-β1 were found in PAD patients (ankle-brachial index, ABI⩽0.9) compared to non-PAD control subjects (1.4>ABI>0.9). CONCLUSION The results demonstrated the subjects with unrecognized PAD (ABI⩽0.9) show a characteristic phlogistic pattern differently from healthy subjects and it strongly supports the pivotal role played by inflammatory and immunological mechanisms in the initiation and progression of the atherosclerotic process in peripheral arteries. These biomarkers could be helpful to screen the susceptibility for the diseases in peripheral arteries.
Collapse
|
39
|
Evaluation of serum adipokines in peripheral arterial occlusive disease. Mediators Inflamm 2012; 2012:257808. [PMID: 22547903 PMCID: PMC3324910 DOI: 10.1155/2012/257808] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 01/06/2012] [Accepted: 01/20/2012] [Indexed: 01/29/2023] Open
Abstract
Aim. Out study aimed to assess the serum levels of adipokines in patients with peripheral arterial occlusive disease (PAOD) caused by atherosclerosis. Methods. Serum samples were obtained from 221 patients. One hundred and forty patients, (26 females and 114 males) met the inclusion criteria and were assigned into the case group. Eighty one patients (17 females and 64 males), were included in the control group. Circulating plasma levels of adiponectin, leptin, resistin, and TNF-α were measured using the enzyme-linked immunosorbent assay (ELISA) method. Results. Significant lower levels of adiponectin were present (P = 0.0061) in PAOD patients (2380.23 ± 1634.42 pg/mL) compared to the control group (3065.06 ± 1901.2 pg/mL). The mean value of leptin (2844.42 ± 3301.08 pg/mL) and resistin (2047.81±3301.08 pg/mL) patients included in the PAOD group was higher, as compared to the control group. Statistically significant difference was found between the two groups for leptin (P = 0.0332) and for resistin (P = 0.0352). No statistically significant difference for TNF-α was found between the two groups (P > 0.05). Conclusion. The markers of inflammation secreted by the adipose tissue (adiponectin, leptin, resistin) showed significant differences in patients from the case group (with PAOD) compared to the control group.
Collapse
|
40
|
Burgess A, Vanella L, Bellner L, Schwartzman ML, Abraham NG. Epoxyeicosatrienoic acids and heme oxygenase-1 interaction attenuates diabetes and metabolic syndrome complications. Prostaglandins Other Lipid Mediat 2012; 97:1-16. [PMID: 22100745 PMCID: PMC3261364 DOI: 10.1016/j.prostaglandins.2011.10.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 10/05/2011] [Accepted: 10/17/2011] [Indexed: 12/19/2022]
Abstract
MSCs are considered to be the natural precursors to adipocyte development through the process of adipogenesis. A link has been established between decreased protective effects of EETs or HO-1 and their interaction in metabolic syndrome. Decreases in HO-1 or EET were associated with an increase in adipocyte stem cell differentiation and increased levels of inflammatory cytokines. EET agonist (AKR-I-27-28) inhibited MSC-derived adipocytes and decreased the levels of inflammatory cytokines. We further describe the role of CYP-epoxygenase expression, HO expression, and circulating cytokine levels in an obese mouse, ob/ob(-/-) mouse model. Ex vivo measurements of EET expression within MSCs derived from ob/ob(-/-) showed decreased levels of EETs that were increased by HO induction. This review demonstrates that suppression of HO and EET systems exist in MSCs prior to the development of adipocyte dysfunction. Further, adipocyte dysfunction can be ameliorated by induction of HO-1 and CYP-epoxygenase, i.e. EET.
Collapse
Affiliation(s)
- Angela Burgess
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, OH 43614
| | - Luca Vanella
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, OH 43614
| | - Lars Bellner
- Department of Pharmacology, New York Medical College, Valhalla, NY, 10595
| | | | - Nader G. Abraham
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, OH 43614
| |
Collapse
|
41
|
Niida T, Isoda K, Kitagaki M, Ishigami N, Adachi T, Matsubara O, Takeda K, Kishimoto T, Ohsuzu F. IκBNS regulates interleukin-6 production and inhibits neointimal formation after vascular injury in mice. Cardiovasc Res 2011; 93:371-9. [PMID: 22135163 DOI: 10.1093/cvr/cvr323] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
AIMS IκBNS regulates a subset of Toll-like receptor (TLR)-dependent genes including interleukin-6 (IL-6) by inhibiting nuclear factor-κB (NF-κB). IL-6 is an inflammatory biomarker for cardiovascular diseases. The aim of this study was to determine whether IκBNS changes arterial inflammation and intimal hyperplasia after vascular injury. METHODS AND RESULTS We investigated neointimal formation in IκBNS-deficient (IκBNS(-/-); C57BL/6 background) and wild-type (IκBNS(+/+)) mice 2 weeks after cuff injury. The mean intimal area and the intima/media ratio of IκBNS(-/-) mice increased 89% (8066 ± 1141 vs. 4267 ± 1095 μm(2); P = 0.027) and 100% (0.72 ± 0.13 vs. 0.36 ± 0.09; P = 0.032) compared with IκBNS(+/+) mice. We observed significant up-regulation of TLR4 in injured arteries of IκBNS(-/-) mice. NF-κB activity in the intima of IκBNS(-/-) mice was 5.1-fold higher (P = 0.008) compared with IκBNS(+/+) mice at 7 days post-injury. IL-6 mRNA levels in injured arteries of IκBNS(-/-) mice were 1.8-fold higher (P = 0.002) compared with those of IκBNS(+/+) mice at 3 days post-injury. Vascular smooth muscle cells from IκBNS(-/-) mice showed a significant increase in cell migration compared with those from IκBNS(+/+) mice after IL-6 stimulation in the scratch-wound healing assay. Furthermore, anti-mouse IL-6 receptor antibody (MR16-1) significantly reduced intimal hyperplasia compared with control IgG injection in IκBNS(-/-) mice. These findings suggest that IL-6 participates in the development of neointimal hyperplasia after vascular injury in IκBNS(-/-) mice. CONCLUSION IκBNS down-regulates TLR4 expression, NF-κB activity, and IL-6 production after vascular injury. IκBNS might suppress intimal hyperplasia caused by vascular inflammation such as atherosclerosis, and restenosis after angioplasty.
Collapse
Affiliation(s)
- Tomiharu Niida
- Department of Internal Medicine 1, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Singh T, Newman AB. Inflammatory markers in population studies of aging. Ageing Res Rev 2011; 10:319-29. [PMID: 21145432 DOI: 10.1016/j.arr.2010.11.002] [Citation(s) in RCA: 639] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 11/15/2010] [Accepted: 11/18/2010] [Indexed: 01/04/2023]
Abstract
PURPOSE To review findings from major epidemiologic studies regarding risk factors for and consequences of elevated markers of inflammation in older adults. RESULTS Most large, current epidemiologic studies of older adults have measured serum interleukin-6 (IL-6), C-reactive protein (CRP) and tumor necrosis factor alpha (TNF-alpha) and some studies also include more extensive batteries of measures including soluble receptors. There are few defined risk factors for the modest elevations in inflammatory markers seen with aging. These include visceral adiposity, lower sex steroid hormones, smoking, depression and periodontal disease. Of the markers assessed, IL-6 is most robustly associated with incident disease, disability and mortality. CONCLUSION Though correlated with age, the etiology of elevated inflammatory markers remains incompletely defined. Inflammation, especially IL-6 may be a common cause of multiple age-related diseases or a final common pathway by which disease leads to disability and adverse outcomes in older adults. Future research targeting inflammation should examine these pathways.
Collapse
Affiliation(s)
- Tushar Singh
- University of Pittsburgh, Graduate School of Public Health, Department of Epidemiology, Pittsburgh, PA, USA
| | | |
Collapse
|
43
|
Lau JF, Weinberg MD, Olin JW. Peripheral artery disease. Part 1: clinical evaluation and noninvasive diagnosis. Nat Rev Cardiol 2011; 8:405-18. [PMID: 21629211 DOI: 10.1038/nrcardio.2011.66] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Peripheral artery disease (PAD) is a marker of systemic atherosclerosis. Most patients with PAD also have concomitant coronary artery disease (CAD), and a large burden of morbidity and mortality in patients with PAD is related to myocardial infarction, ischemic stroke, and cardiovascular death. PAD patients without clinical evidence of CAD have the same relative risk of death from cardiac or cerebrovascular causes as those diagnosed with prior CAD, consistent with the systemic nature of the disease. The same risk factors that contribute to CAD and cerebrovascular disease also lead to the development of PAD. Because of the high prevalence of asymptomatic disease and because only a small percentage of PAD patients present with classic claudication, PAD is frequently underdiagnosed and thus undertreated. Health care providers may have difficulty differentiating PAD from other diseases affecting the limb, such as arthritis, spinal stenosis or venous disease. In Part 1 of this Review, we explain the epidemiology of and risk factors for PAD, and discuss the clinical presentation and diagnostic evaluation of patients with this condition.
Collapse
Affiliation(s)
- Joe F Lau
- The Zena and Michael A. Wiener Cardiovascular Institute, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1033, New York, NY 10029-6574, USA
| | | | | |
Collapse
|
44
|
Tsai PC, Lin TH, Hsu PC, Wang YS, Liao YC, Juo SHH. Polymorphism of 270 A > G in BRAP is Associated with Lower Ankle-Brachial Index in a Taiwanese Population. J Atheroscler Thromb 2011; 18:413-20. [DOI: 10.5551/jat.7468] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Pei-Chien Tsai
- Department of Medical Research, Kaohsiung Medical University Hospital
| | - Tsung-Hsien Lin
- Division of Cardiology, Kaohsiung Medical University Hospital
- Department of Internal Medicine, Kaohsiung Medical University
| | - Po-Chao Hsu
- Division of Cardiology, Kaohsiung Medical University Hospital
| | - Yung-Song Wang
- Department of Medical Research, Kaohsiung Medical University Hospital
| | - Yi-Chu Liao
- Graduate Institute of Medicine, Kaohsiung Medical University
- Section of Neurology, Taichung Veterans General Hospital
| | - Suh-Hang Hank Juo
- Department of Medical Research, Kaohsiung Medical University Hospital
- Department of Medical Genetics, Kaohsiung Medical University
| |
Collapse
|
45
|
Burgess A, Li M, Vanella L, Kim DH, Rezzani R, Rodella L, Sodhi K, Canestraro M, Martasek P, Peterson SJ, Kappas A, Abraham NG. Adipocyte heme oxygenase-1 induction attenuates metabolic syndrome in both male and female obese mice. Hypertension 2010; 56:1124-30. [PMID: 21041703 DOI: 10.1161/hypertensionaha.110.151423] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Increases in visceral fat are associated with increased inflammation, dyslipidemia, insulin resistance, glucose intolerance, and vascular dysfunction. We examined the effect of the potent heme oxygenase (HO)-1 inducer, cobalt protoporphyrin (CoPP), on regulation of adiposity and glucose levels in both female and male obese mice. Both lean and obese mice were administered CoPP intraperitoneally (3 mg/kg once per week) for 6 weeks. Serum levels of adiponectin, tumor necrosis factor α (TNFa), interleukin (IL)-1β and IL-6, and HO-1, PPARγ, pAKT, and pAMPK protein expression in adipocytes and vascular tissue were measured. While female obese mice continued to gain weight at a rate similar to controls, induction of HO-1 slowed the rate of weight gain in male obese mice. HO-1 induction led to lowered blood pressure levels in obese male and female mice similar to that of lean male and female mice. HO-1 induction also produced a significant decrease in the plasma levels of IL-6, TNFα, IL-1β, and fasting glucose of obese females compared to untreated female obese mice. HO-1 induction increased the number and decreased the size of adipocytes of obese animals. HO-1 induction increased adiponectin, pAKT, pAMPK, and PPARγ levels in adipocyte of obese animals. Induction of HO-1 in adipocytes was associated with an increase in adiponectin and a reduction in inflammatory cytokines. These findings offer the possibility of treating not only hypertension, but also other detrimental metabolic consequences of obesity including insulin resistance and dyslipidemia in obese populations by induction of HO-1 in adipocytes.
Collapse
Affiliation(s)
- Angela Burgess
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Petrescu F, Voican SC, Silosi I. Tumor necrosis factor-alpha serum levels in healthy smokers and nonsmokers. Int J Chron Obstruct Pulmon Dis 2010; 5:217-22. [PMID: 20714375 PMCID: PMC2921689 DOI: 10.2147/copd.s8330] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Indexed: 11/23/2022] Open
Abstract
Background: Tobacco smoking is the most important risk factor for chronic obstructive pulmonary disease (COPD) development. Inhaled cigarette smoke can induce tumor necrosis factor-α (TNF-α) production by alveolar macrophages, which in turn may enhance the production of metalloproteinases (MMPs). MMPs have been involved in mediating airway inflammation and lung destruction. Objectives: We aimed to measure the TNF-α serum levels in healthy heavy smokers and healthy nonsmokers to determine the dose-response relationship based on the cigarette smoke exposure. Subjects and methods: We included in our study 43 healthy heavy smokers and 19 healthy nonsmokers (the control group). The smokers group was classified as less than one pack, one pack, and more than one pack per day. A clinical and paraclinical evaluation was performed in both groups, without any evidence of infection or COPD. The serum levels of TNF-α were assessed by ELISA. Results: The TNF-α serum levels were significantly higher for the group of smokers compared to the group of nonsmokers (P < 0.004). We also noticed an increased TNF-α concentration in the serum of smokers with more than one pack per day compared with those with less than one pack per day (P < 0.03). There was a positive correlation between the serum level of TNF-α and tobacco smoke exposure. Conclusions: The high levels of TNF-α in the serum of smokers suggest an imbalance between the proinflammatory and anti-inflammatory factors as a result of tobacco smoke exposure. The concentration of TNF-α is elevated in the serum of healthy heavy smokers in a cigarette dose-dependent manner. We speculate that the serum level of TNF-α might be a useful biomarker for the selection of heavy smokers with a high risk of developing smoke induced pulmonary diseases.
Collapse
Affiliation(s)
- Florin Petrescu
- Department of Internal Medicine, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | | | | |
Collapse
|
47
|
Cooke JP, Wilson AM. Biomarkers of peripheral arterial disease. J Am Coll Cardiol 2010; 55:2017-23. [PMID: 20447524 DOI: 10.1016/j.jacc.2009.08.090] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2009] [Revised: 07/22/2009] [Accepted: 08/16/2009] [Indexed: 10/19/2022]
Abstract
Atherosclerotic arterial occlusive disease affecting the lower extremities is also known as peripheral artery disease (PAD). This disorder affects 8 to 12 million individuals in the U.S. and is increasingly prevalent in Europe and Asia. Unfortunately, most patients are not diagnosed and are not optimally treated. A blood test for PAD, if sufficiently sensitive and specific, would be expected to improve recognition and treatment of these individuals. Even a biomarker panel of moderate sensitivity and specificity for PAD could refine risk stratification to select individuals for diagnostic vascular examination. Alternatively, biomarkers for PAD may be useful in determining prognosis, the risk for progression, or the response to therapy. Finally, the discovery of biomarkers associated with PAD may provide novel insights into the pathophysiology of PAD and new therapeutic avenues to pursue. Biomarkers may be derived from studies of the genome, transcriptome, proteome, or metabolome. The focus of this review is on proteomic biomarkers associated with PAD.
Collapse
Affiliation(s)
- John P Cooke
- Division of Cardiovascular Medicine, Stanford University, Stanford, California 94305-5406, USA.
| | | |
Collapse
|
48
|
Utilidad clínica del índice tobillo-brazo en el paciente con enfermedad coronaria. Med Clin (Barc) 2010; 134:208-10. [DOI: 10.1016/j.medcli.2009.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Accepted: 09/07/2009] [Indexed: 11/19/2022]
|
49
|
Kim SH, Jeong MH, Lee MG, Ko JS, Park KH, Sim DS, Hong YJ, Kim JH, Ahn Y, Kang JC. The Relationship between the Levels of Lipoprotein (a) and Fibrinogen and Clinical Outcome in Patients with Acute Myocardial Infarction. Chonnam Med J 2010. [DOI: 10.4068/cmj.2010.46.1.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Su Hyun Kim
- The Heart Center of Chonnam National University Hospital, Heart Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health, Welfare and Family Affairs, Gwangju, Korea
| | - Myung Ho Jeong
- The Heart Center of Chonnam National University Hospital, Heart Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health, Welfare and Family Affairs, Gwangju, Korea
| | - Min Goo Lee
- The Heart Center of Chonnam National University Hospital, Heart Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health, Welfare and Family Affairs, Gwangju, Korea
| | - Jum Suk Ko
- The Heart Center of Chonnam National University Hospital, Heart Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health, Welfare and Family Affairs, Gwangju, Korea
| | - Keun-Ho Park
- The Heart Center of Chonnam National University Hospital, Heart Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health, Welfare and Family Affairs, Gwangju, Korea
| | - Doo Sun Sim
- The Heart Center of Chonnam National University Hospital, Heart Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health, Welfare and Family Affairs, Gwangju, Korea
| | - Young Joon Hong
- The Heart Center of Chonnam National University Hospital, Heart Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health, Welfare and Family Affairs, Gwangju, Korea
| | - Ju Han Kim
- The Heart Center of Chonnam National University Hospital, Heart Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health, Welfare and Family Affairs, Gwangju, Korea
| | - Youngkeun Ahn
- The Heart Center of Chonnam National University Hospital, Heart Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health, Welfare and Family Affairs, Gwangju, Korea
| | - Jung Chaee Kang
- The Heart Center of Chonnam National University Hospital, Heart Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health, Welfare and Family Affairs, Gwangju, Korea
| |
Collapse
|
50
|
Morillas P, Quiles J, Cordero A, Guindo J, Soria F, Mazón P, Gonzalez-Juanatey JR, Bertomeu V. Impact of clinical and subclinical peripheral arterial disease in mid-term prognosis of patients with acute coronary syndrome. Am J Cardiol 2009; 104:1494-8. [PMID: 19932781 DOI: 10.1016/j.amjcard.2009.07.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2009] [Revised: 07/02/2009] [Accepted: 07/02/2009] [Indexed: 01/22/2023]
Abstract
Observational studies report poor prognosis of patients after acute coronary syndrome (ACS) in the presence of previous peripheral arterial disease (PAD), but data on subclinical PAD are scarce. This study was designed to assess the predictive value of clinical and subclinical PAD in the follow-up of patients after an ACS. We included 1,054 patients hospitalized for an ACS who survived the acute phase. Patients were divided into 3 groups: clinical PAD (previously diagnosed PAD or intermittent claudication), subclinical PAD (defined as ankle-brachial index <or=0.9 or >1.4), and no PAD. Clinical PAD was present in 150 patients (14.2%) and 298 cases of subclinical PAD were detected (28.3%). Patients with PAD (clinical and subclinical PAD) were significantly older and had a higher prevalence of hypertension and diabetes mellitus than those without PAD. During the 1-year follow-up, 59 patients died (5.6%). Previous PAD (hazard ratio 4.38, 95% confidence interval 1.96 to 9.82, p <0.001) and subclinical PAD (hazard ratio 2.35, 95% confidence interval 1.05 to 5.23, p <0.05) were associated with increased cardiovascular mortality. Moreover, patients with clinical PAD had higher rates of major cardiovascular events (myocardial infarction, angina, and heart failure) than patients with subclinical PAD or without PAD. In conclusion, beyond clinical PAD, measurement of ankle-brachial index after ACS provides substantial information on intermediate-term prognosis.
Collapse
|