1
|
Bello MO, Wadid M, Malode A, Patel V, Shah A, Vyas A, Ahmad HA, Tarun T, Dani S, Ahmad J, Zarwan C, Ganatra S. Atrial Fibrillation in Patients with Breast Cancer: A Literature Review. Cardiol Ther 2025; 14:1-15. [PMID: 39714744 PMCID: PMC11893935 DOI: 10.1007/s40119-024-00394-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024] Open
Abstract
In addition to traditional risk factors, patients with breast cancer are at an increased risk of atrial fibrillation due to cancer itself and certain cancer therapies. Atrial fibrillation in these patients adds to their morbidity and mortality. The precise mechanisms leading to the increased atrial fibrillation in patients with breast cancer are not well understood. The main goal of atrial fibrillation management in this population is to facilitate uninterrupted cancer treatment while addressing the arrhythmia and other cardiovascular sequelae of cancer treatment. Rhythm control is often challenging to implement in patients with breast cancer during active antineoplastic therapy because of the need for uninterrupted anticoagulation, potential drug-drug interactions between cancer treatments and antiarrhythmic medications, and the increased likelihood of atrial fibrillation recurrence. Prevention of thromboembolism and anticoagulation can also be challenging in patients with breast cancer as a result of the increased risk of cancer-related procoagulant state and coagulopathies. The integration of a cardio-oncology team and a multidisciplinary approach are crucial for better outcomes. The therapeutic interventions should be tailored toward individual patients' profiles through a shared decision-making approach. The precise mechanisms leading to the increased atrial fibrillation in patients with breast cancer are not well understood, highlighting the gaps in our knowledge. More research is required to reduce these gaps, refine risk stratification, and optimize treatment strategies in these patients.
Collapse
Affiliation(s)
- Mozidat Olamide Bello
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Mark Wadid
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Aishwarya Malode
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Vahin Patel
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Anuj Shah
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Ankit Vyas
- Department of Vascular Medicine, Ochsner Clinic Foundation, New Orleans, LA, USA
| | | | - Tushar Tarun
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sourbha Dani
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Javaria Ahmad
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Corrine Zarwan
- Division of Hematology/Oncology, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA.
| |
Collapse
|
2
|
Dungan GD, Kantarcioglu B, Odeh A, Hoppensteadt D, Siddiqui F, Rohde L, Fareed J, Syed MA. Vascular Endothelial Dysfunction and Immunothrombosis in the Pathogenesis of Atrial Fibrillation. Clin Appl Thromb Hemost 2024; 30:10760296241296138. [PMID: 39654486 PMCID: PMC11629412 DOI: 10.1177/10760296241296138] [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: 03/06/2024] [Revised: 10/07/2024] [Accepted: 10/14/2024] [Indexed: 12/12/2024] Open
Abstract
Atrial Fibrillation (AF) induces proinflammatory processes which incite vascular endothelial activation and dysfunction. This study seeks to examine the potential relationship between various endothelial, inflammatory, thrombotic, and renin-angiotensin-system (RAS) biomarkers in AF patients.Blood samples were from AF patients (n = 110) prospectively enrolled in this study prior to their first AF ablation. Control plasma samples (n = 100) were used as reference. All samples were analyzed for endothelial (NO, ICAM-1, VEGF, TF, TFPI, TM, Annexin V), inflammatory (IL-6, TNFα, CRP), thrombotic (vWF, tPA, PAI-1, TAFI, D-dimer), and RAS (Renin, Ang-II) biomarkers using ELISA methods. Biomarker average comparisons and Spearman correlations were performed.AF patients showed varying levels of biomarker increase compared to controls. We observed a significant decrease of Ang-II in the AF population relative to controls when stratified for the use of angiotensin-converting enzyme inhibitor (ACEI) or angiotensin II receptor blocker (ARB) upon study enrollment. AF patients showed statistically significant correlations between the following biomarkers: TNFα vs IL-6 (rs = 0.317, p = .004), ICAM-1 vs TNFα (rs = 0.527, p = .012), Annexin V vs VEGF (rs = 0.620, p < .001), CRP vs VEGF (rs = 0.342, p = .031), Ang-II vs tPA (rs = -0.592, p = .010), and tPA vs PAI-1 (rs = 0.672, p < .001).Our study demonstrated significant elevation of endothelial, inflammatory, and thrombotic biomarkers in AF patients compared to controls, with significant correlations between these biomarkers in the AF population. Future investigations are required to better elucidate the mechanistic pathways that lead to endothelial dysfunction and thromboinflammation in AF. This may provide novel therapeutic targets, that in addition to current anticoagulation practices, can best curtail thrombogenicity in AF.
Collapse
Affiliation(s)
- Gabriel D. Dungan
- Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Bulent Kantarcioglu
- Department of Pathology and Laboratory Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - Ameer Odeh
- Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Debra Hoppensteadt
- Department of Pathology and Laboratory Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - Fakiha Siddiqui
- Department of Pathology and Laboratory Medicine, Loyola University Medical Center, Maywood, IL, USA
- Program in Health Sciences, UCAM- Universidad Católica San Antonio de Murcia, Murcia, Spain
| | - Luke Rohde
- Department of Pathology and Laboratory Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - Jawed Fareed
- Department of Pathology and Laboratory Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - Mushabbar A. Syed
- Department of Medicine, Division of Cardiology, Loyola University Medical Center, Maywood, IL, USA
| |
Collapse
|
3
|
Procyk G, Bilicki D, Balsam P, Lodziński P, Grabowski M, Gąsecka A. Extracellular Vesicles in Atrial Fibrillation—State of the Art. Int J Mol Sci 2022; 23:ijms23147591. [PMID: 35886937 PMCID: PMC9325220 DOI: 10.3390/ijms23147591] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 11/20/2022] Open
Abstract
Extracellular vesicles are particles released from cells and delimited by a lipid bilayer. They have been widely studied, including extensive investigation in cardiovascular diseases. Many scientists have explored their role in atrial fibrillation. Patients suffering from atrial fibrillation have been evidenced to present altered levels of these particles as well as changed amounts of their contents such as micro-ribonucleic acids (miRs). Although many observations have been made so far, a large randomized clinical trial is needed to assess the previous findings. This review aims to thoroughly summarize current research regarding extracellular vesicles in atrial fibrillation.
Collapse
Affiliation(s)
- Grzegorz Procyk
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; (P.B.); (P.L.); (M.G.); (A.G.)
- Correspondence: ; Tel.: +48-723-488-305
| | - Dominik Bilicki
- Faculty of Medicine, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland;
| | - Paweł Balsam
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; (P.B.); (P.L.); (M.G.); (A.G.)
| | - Piotr Lodziński
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; (P.B.); (P.L.); (M.G.); (A.G.)
| | - Marcin Grabowski
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; (P.B.); (P.L.); (M.G.); (A.G.)
| | - Aleksandra Gąsecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; (P.B.); (P.L.); (M.G.); (A.G.)
| |
Collapse
|
4
|
Xiang K, Akram M, Elbossaty WF, Yang J, Fan C. Exosomes in atrial fibrillation: therapeutic potential and role as clinical biomarkers. Heart Fail Rev 2022; 27:1211-1221. [PMID: 34251579 DOI: 10.1007/s10741-021-10142-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/05/2021] [Indexed: 12/21/2022]
Abstract
Atrial fibrillation (AF), the most common cardiac arrhythmia, is a global epidemic. AF can cause heart failure and myocardial infarction and increase the risk of stroke, disability, and thromboembolic events. AF is becoming increasingly ubiquitous and is associated with increased morbidity and mortality at higher ages, resulting in an increasing threat to human health as well as substantial medical and social costs. Currently, treatment strategies for AF focus on controlling heart rate and rhythm with medications to restore and maintain sinus rhythm, but this approach has limitations. Catheter ablation is not entirely satisfactory and does not address the issues underlying AF. Research exploring the mechanisms causing AF is urgently needed for improved prevention, diagnosis, and treatment of AF. Exosomes are small vesicles (30-150 nm) released by cells that transmit information between cells. MicroRNAs in exosomes play an important role in the pathogenesis of AF and are established as a biomarker for AF. In this review, a summary of the role of exosomes in AF is presented. The role of exosomes and microRNAs in AF occurrence, their therapeutic potential, and their potential role as clinical biomarkers is considered. A better understanding of exosomes has the potential to improve the prognosis of AF patients worldwide, reducing the global medical burden of this disease.
Collapse
Affiliation(s)
- Kun Xiang
- Department of Cardiovascular Surgery, the Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Muhammad Akram
- Department of Eastern Medicine, Government College University Faisalabad, Faisalabad, Pakistan
| | | | - Jinfu Yang
- Department of Cardiovascular Surgery, the Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, the Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China.
| |
Collapse
|
5
|
Berezin AE, Berezin AA. Extracellular Vesicles and Thrombogenicity in Atrial Fibrillation. Int J Mol Sci 2022; 23:1774. [PMID: 35163695 PMCID: PMC8836440 DOI: 10.3390/ijms23031774] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 01/30/2022] [Accepted: 02/02/2022] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) are defined as a heterogenic group of lipid bilayer vesicular structures with a size in the range of 30-4000 nm that are released by all types of cultured cells. EVs derived from platelets, mononuclears, endothelial cells, and adipose tissue cells significantly increase in several cardiovascular diseases, including in atrial fibrillation (AF). EVs are engaged in cell-to-cell cooperation, endothelium integrity, inflammation, and immune response and are a cargo for several active molecules, such as regulatory peptides, receptors, growth factors, hormones, and lipids. Being transductors of the intercellular communication, EVs regulate angiogenesis, neovascularization, coagulation, and maintain tissue reparation. There is a large amount of evidence regarding the fact that AF is associated with elevated levels of EVs derived from platelets and mononuclears and a decreased number of EVs produced by endothelial cells. Moreover, some invasive procedures that are generally performed for the treatment of AF, i.e., pulmonary vein isolation, were found to be triggers for elevated levels of platelet and mononuclear EVs and, in turn, mediated the transient activation of the coagulation cascade. The review depicts the role of EVs in thrombogenicity in connection with a risk of thromboembolic complications, including ischemic stroke and systemic thromboembolism, in patients with various forms of AF.
Collapse
Affiliation(s)
- Alexander E. Berezin
- Internal Medicine Department, State Medical University, Zaporozhye 69035, Ukraine
| | - Alexander A. Berezin
- Internal Medicine Department, Medical Academy of Postgraduate Education, Zaporozhye 69096, Ukraine;
| |
Collapse
|
6
|
Siwaponanan P, Kaewkumdee P, Phromawan W, Udompunturak S, Chomanee N, Udol K, Pattanapanyasat K, Krittayaphong R. Increased expression of six-large extracellular vesicle-derived miRNAs signature for nonvalvular atrial fibrillation. J Transl Med 2022; 20:4. [PMID: 34980172 PMCID: PMC8722074 DOI: 10.1186/s12967-021-03213-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/19/2021] [Indexed: 12/17/2022] Open
Abstract
Backgrounds Non-valvular atrial fibrillation (AF) is the most common type of cardiac arrhythmia. AF is caused by electrophysiological abnormalities and alteration of atrial tissues, which leads to the generation of abnormal electrical impulses. Extracellular vesicles (EVs) are membrane-bound vesicles released by all cell types. Large EVs (lEVs) are secreted by the outward budding of the plasma membrane during cell activation or cell stress. lEVs are thought to act as vehicles for miRNAs to modulate cardiovascular function, and to be involved in the pathophysiology of cardiovascular diseases (CVDs), including AF. This study identified lEV-miRNAs that were differentially expressed between AF patients and non-AF controls. Methods lEVs were isolated by differential centrifugation and characterized by Nanoparticle Tracking Analysis (NTA), Transmission Electron Microscopy (TEM), flow cytometry and Western blot analysis. For the discovery phase, 12 AF patients and 12 non-AF controls were enrolled to determine lEV-miRNA profile using quantitative reverse transcription polymerase chain reaction array. The candidate miRNAs were confirmed their expression in a validation cohort using droplet digital PCR (30 AF, 30 controls). Bioinformatics analysis was used to predict their target genes and functional pathways. Results TEM, NTA and flow cytometry demonstrated that lEVs presented as cup shape vesicles with a size ranging from 100 to 1000 nm. AF patients had significantly higher levels of lEVs at the size of 101–200 nm than non-AF controls. Western blot analysis was used to confirm EV markers and showed the high level of cardiomyocyte expression (Caveolin-3) in lEVs from AF patients. Nineteen miRNAs were significantly higher (> twofold, p < 0.05) in AF patients compared to non-AF controls. Six highly expressed miRNAs (miR-106b-3p, miR-590-5p, miR-339-3p, miR-378-3p, miR-328-3p, and miR-532-3p) were selected to confirm their expression. Logistic regression analysis showed that increases in the levels of these 6 highly expressed miRNAs associated with AF. The possible functional roles of these lEV-miRNAs may involve in arrhythmogenesis, cell apoptosis, cell proliferation, oxygen hemostasis, and structural remodeling in AF. Conclusion Increased expression of six lEV-miRNAs reflects the pathophysiology of AF that may provide fundamental knowledge to develop the novel biomarkers for diagnosis or monitoring the patients with the high risk of AF. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03213-6.
Collapse
Affiliation(s)
- Panjaree Siwaponanan
- Siriraj Center of Research Excellence for Microparticle and Exosome in Diseases, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pontawee Kaewkumdee
- Division of Cardiology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wilasinee Phromawan
- Division of Cardiology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Suthipol Udompunturak
- Division of Clinical Epidemiology, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nusara Chomanee
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kamol Udol
- Department of Preventive and Social Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kovit Pattanapanyasat
- Siriraj Center of Research Excellence for Microparticle and Exosome in Diseases, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Rungroj Krittayaphong
- Division of Cardiology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
7
|
Etli M. Investigation of routine blood parameters for predicting embolic risk in patients with nonvalvular atrial fibrillation. INDIAN JOURNAL OF VASCULAR AND ENDOVASCULAR SURGERY 2022. [DOI: 10.4103/ijves.ijves_77_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
8
|
Black N, Mohammad F, Saraf K, Morris G. Endothelial function and atrial fibrillation: A missing piece of the puzzle? J Cardiovasc Electrophysiol 2021; 33:109-116. [PMID: 34674346 DOI: 10.1111/jce.15277] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 09/14/2021] [Accepted: 10/18/2021] [Indexed: 12/15/2022]
Abstract
Endothelial dysfunction, a term used to describe both the physical damage and dysregulated physiology of this endothelial lining, is an increasingly recognized pathophysiological state shared by many cardiovascular diseases. Historically, the role of endothelial dysfunction in atrial fibrillation (AF) was thought to be limited to mediating atrial thromboembolism. However, there is emerging evidence that endothelial dysfunction both promotes and maintains atrial arrhythmic substrate, predicts adverse outcomes, and identifies patients at high risk of recurrence following cardioversion and ablation therapy. Treatments targeted at improving endothelial function also represent a promising new therapeutic paradigm in AF. This review summarizes the current understanding of endothelial function in AF.
Collapse
Affiliation(s)
- Nicholas Black
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Fahad Mohammad
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Karan Saraf
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Gwilym Morris
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK.,Manchester Heart Centre, Manchester Academic Health Science Centre, Manchester University Foundation Trust, Manchester, UK
| |
Collapse
|
9
|
Weiss L, Keaney J, Szklanna PB, Prendiville T, Uhrig W, Wynne K, Kelliher S, Ewins K, Comer SP, Egan K, O'Rourke E, Moran E, Petrov G, Patel A, Lennon Á, Blanco A, Kevane B, Murphy S, Ní Áinle F, Maguire PB. Nonvalvular atrial fibrillation patients anticoagulated with rivaroxaban compared with warfarin exhibit reduced circulating extracellular vesicles with attenuated pro-inflammatory protein signatures. J Thromb Haemost 2021; 19:2583-2595. [PMID: 34161660 DOI: 10.1111/jth.15434] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 06/02/2021] [Accepted: 06/22/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Rivaroxaban, a direct oral factor Xa inhibitor, mediates anti-inflammatory and cardiovascular-protective effects besides its well-established anticoagulant properties; however, these remain poorly characterized. Extracellular vesicles (EVs) are important circulating messengers regulating a myriad of biological and pathological processes and may be highly relevant to the pathophysiology of atrial fibrillation as they reflect alterations in platelet and endothelial biology. However, the effects of rivaroxaban on circulating pro-inflammatory EVs remain unknown. OBJECTIVES We hypothesized that rivaroxaban's anti-inflammatory properties are reflected upon differential molecular profiles of circulating EVs. METHODS Differences in circulating EV profiles were assessed using a combination of single vesicle analysis by Nanoparticle Tracking Analysis and flow cytometry, and proteomics. RESULTS We demonstrate, for the first time, that rivaroxaban-treated non-valvular atrial fibrillation (NVAF) patients (n=8) exhibit attenuated inflammation compared with matched warfarin controls (n=15). Circulating EV profiles were fundamentally altered. Moreover, quantitative proteomic analysis of enriched plasma EVs from six pooled biological donors per treatment group revealed a profound decrease in highly pro-inflammatory protein expression and complement factors, together with increased expression of negative regulators of inflammatory pathways. Crucially, a reduction in circulating levels of soluble P-selectin was observed in rivaroxaban-treated patients (compared with warfarin controls), which negatively correlated with the patient's time on treatment. CONCLUSION Collectively, these data demonstrate that NVAF patients anticoagulated with rivaroxaban (compared with warfarin) exhibit both a reduced pro-inflammatory state and evidence of reduced endothelial activation. These findings are of translational relevance toward characterizing the anti-inflammatory and cardiovascular-protective mechanisms associated with rivaroxaban therapy.
Collapse
Affiliation(s)
- Luisa Weiss
- UCD Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - John Keaney
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Cardiology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Paulina B Szklanna
- UCD Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Tadhg Prendiville
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Wido Uhrig
- UCD Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
| | - Kieran Wynne
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Sarah Kelliher
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Karl Ewins
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Shane P Comer
- UCD Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Karl Egan
- UCD Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Ellen O'Rourke
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Eric Moran
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Georgi Petrov
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Ashish Patel
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Áine Lennon
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Alfonso Blanco
- Flow Cytometry Core, Conway Institute, University College Dublin, Dublin, Ireland
| | - Barry Kevane
- UCD Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Sean Murphy
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Stroke Medicine, Mater Misericordiae University Hospital, Dublin, Ireland
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Fionnuala Ní Áinle
- UCD Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Patricia B Maguire
- UCD Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- UCD Institute for Discovery, O'Brien Centre for Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
10
|
Lenart-Migdalska A, Drabik L, Kaźnica-Wiatr M, Tomkiewicz-Pająk L, Podolec P, Olszowska M. Flow Cytometric Assessment of Endothelial and Platelet Microparticles in Patients With Atrial Fibrillation Treated With Dabigatran. Clin Appl Thromb Hemost 2021; 26:1076029620972467. [PMID: 33237804 PMCID: PMC7787695 DOI: 10.1177/1076029620972467] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The prothrombotic state in patients with atrial fibrillation (AF) is related to
endothelial injury, the activation of platelets and the coagulation cascade. We
evaluated the levels of platelet- (CD42b) and endothelial-derived (CD144)
microparticles in the plasma patients with non-valvular AF treated with
dabigatran at the time of expected minimum and maximum drug plasma
concentrations. Following that, we determined the peak dabigatran plasma
concentration (cpeak ). CD42b increased after taking dabigatran
(median [IQR] 36.7 [29.4-53.3] vs. 45.6 [32.3-59.5] cells/µL; p = 0.025). The
concentration of dabigatran correlated negatively with the post-dabigatran
change in CD42b (ΔCD42b, r = -0.47, p = 0.021). In the multivariate model, the
independent predictors of ΔCD42b were: cpeak (HR -0.55; with a 95%
confidence interval, CI [-0.93, -0.16]; p = 0.007), coronary artery disease
(CAD) (HR -0.41; 95% CI [-0.79, -0.02]; p = 0.037) and peripheral artery disease
(PAD) (HR 0.42; 95% CI [0.07, 0.74]; p = 0.019). CD144 did not increase after
dabigatran administration. These data suggest that low concentrations of
dabigatran may be associated with platelet activation. PAD and CAD have distinct
effects on CD42b levels during dabigatran treatment.
Collapse
Affiliation(s)
- Aleksandra Lenart-Migdalska
- Department of Cardiac and Vascular Diseases, Institute of Cardiology, Faculty of Medicine, Jagiellonian University Medical College, John Paul II Hospital, Cracow, Poland
| | - Leszek Drabik
- Department of Cardiac and Vascular Diseases, Institute of Cardiology, Faculty of Medicine, Jagiellonian University Medical College, John Paul II Hospital, Cracow, Poland.,Department of Pharmacology, Jagiellonian University Medical College, Cracow, Poland
| | - Magdalena Kaźnica-Wiatr
- Department of Cardiac and Vascular Diseases, Institute of Cardiology, Faculty of Medicine, Jagiellonian University Medical College, John Paul II Hospital, Cracow, Poland
| | - Lidia Tomkiewicz-Pająk
- Department of Cardiac and Vascular Diseases, Institute of Cardiology, Faculty of Medicine, Jagiellonian University Medical College, John Paul II Hospital, Cracow, Poland
| | - Piotr Podolec
- Department of Cardiac and Vascular Diseases, Institute of Cardiology, Faculty of Medicine, Jagiellonian University Medical College, John Paul II Hospital, Cracow, Poland
| | - Maria Olszowska
- Department of Cardiac and Vascular Diseases, Institute of Cardiology, Faculty of Medicine, Jagiellonian University Medical College, John Paul II Hospital, Cracow, Poland
| |
Collapse
|
11
|
Burke H, Wilkinson TMA. Unravelling the mechanisms driving multimorbidity in COPD to develop holistic approaches to patient-centred care. Eur Respir Rev 2021; 30:30/160/210041. [PMID: 34415848 DOI: 10.1183/16000617.0041-2021] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/06/2021] [Indexed: 01/04/2023] Open
Abstract
COPD is a major cause of morbidity and mortality worldwide. Multimorbidity is common in COPD patients and a key modifiable factor, which requires timely identification and targeted holistic management strategies to improve outcomes and reduce the burden of disease.We discuss the use of integrative approaches, such as cluster analysis and network-based theory, to understand the common and novel pathobiological mechanisms underlying COPD and comorbid disease, which are likely to be key to informing new management strategies.Furthermore, we discuss the current understanding of mechanistic drivers to multimorbidity in COPD, including hypotheses such as multimorbidity as a result of shared common exposure to noxious stimuli (e.g. tobacco smoke), or as a consequence of loss of function following the development of pulmonary disease. In addition, we explore the links to pulmonary disease processes such as systemic overspill of pulmonary inflammation, immune cell priming within the inflamed COPD lung and targeted messengers such as extracellular vesicles as a result of local damage as a cause for multimorbidity in COPD.Finally, we focus on current and new management strategies which may target these underlying mechanisms, with the aim of holistic, patient-centred treatment rather than single disease management.
Collapse
Affiliation(s)
- H Burke
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK .,University Hospitals Southampton NHS Foundation Trust, Southampton, UK
| | - T M A Wilkinson
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,University Hospitals Southampton NHS Foundation Trust, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| |
Collapse
|
12
|
Lenart-Migdalska A, Drabik L, Kaźnica-Wiatr M, Tomkiewicz-Pająk L, Podolec P, Olszowska M. Increased Levels of Platelets and Endothelial-Derived Microparticles in Patients With Non-Valvular Atrial Fibrillation During Rivaroxaban Therapy. Clin Appl Thromb Hemost 2021; 27:10760296211019465. [PMID: 34032122 PMCID: PMC8155766 DOI: 10.1177/10760296211019465] [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] [Indexed: 11/16/2022] Open
Abstract
It is known that atrial fibrillation (AF) is associated with the procoagulant
state. Several studies have reported an increase of circulating microparticles
in AF, which may be linked to a hypercoagulable state, atrial thrombosis and
thromboembolism. We evaluated in our study alterations in both platelet (PMP,
CD42b) and endothelial-derived (EMP, CD144) microparticle levels on
anticoagulant therapy with rivaroxaban in nonvalvular AF. After administration
of rivaroxaban, PMP levels were increased (median, [IQR] 35.7 [28.8-47.3] vs.
48.4 [30.9-82.8] cells/µL; P = 0.012), along with an increase
in EMP levels (14.6 [10.0-18.6] vs. 18.3 [12.9-37.1] cells/µL,
P < 0.001). In the multivariable regression analysis,
the independent predictor of post-dose change in PMPs was statin therapy (HR
−0.43; 95% CI −0.75,−0.10, P = 0.011). The post-dose change in
EMPs was also predicted by statin therapy (HR −0.34; 95% CI −0.69, −0.01,
P = 0.046). This study showed an increase in both EMPs and
PMPs at the peak plasma concentration of rivaroxaban. Statins have promising
potential in the prevention of rivaroxaban-related PMP and EMP release. The
pro-thrombotic role of PMPs and EMPs during rivaroxaban therapy requires further
study.
Collapse
Affiliation(s)
- Aleksandra Lenart-Migdalska
- Department of Cardiac and Vascular Diseases, Faculty of Medicine, Jagiellonian University Medical College, Institute of Cardiology, John Paul II Hospital, Kraków, Poland
| | - Leszek Drabik
- Department of Cardiac and Vascular Diseases, Faculty of Medicine, Jagiellonian University Medical College, Institute of Cardiology, John Paul II Hospital, Kraków, Poland.,Department of Pharmacology, Jagiellonian University Medical College, Kraków, Poland
| | - Magdalena Kaźnica-Wiatr
- Department of Cardiac and Vascular Diseases, Faculty of Medicine, Jagiellonian University Medical College, Institute of Cardiology, John Paul II Hospital, Kraków, Poland
| | - Lidia Tomkiewicz-Pająk
- Department of Cardiac and Vascular Diseases, Faculty of Medicine, Jagiellonian University Medical College, Institute of Cardiology, John Paul II Hospital, Kraków, Poland
| | - Piotr Podolec
- Department of Cardiac and Vascular Diseases, Faculty of Medicine, Jagiellonian University Medical College, Institute of Cardiology, John Paul II Hospital, Kraków, Poland
| | - Maria Olszowska
- Department of Cardiac and Vascular Diseases, Faculty of Medicine, Jagiellonian University Medical College, Institute of Cardiology, John Paul II Hospital, Kraków, Poland
| |
Collapse
|
13
|
Abstract
Extracellular vesicles (EVs) have received considerable attention in biological and clinical research due to their ability to mediate cell-to-cell communication. Based on their size and secretory origin, EVs are categorized as exosomes, microvesicles, and apoptotic bodies. Increasing number of studies highlight the contribution of EVs in the regulation of a wide range of normal cellular physiological processes, including waste scavenging, cellular stress reduction, intercellular communication, immune regulation, and cellular homeostasis modulation. Altered circulating EV level, expression pattern, or content in plasma of patients with cardiovascular disease (CVD) may serve as diagnostic and prognostic biomarkers in diverse cardiovascular pathologies. Due to their inherent characteristics and physiological functions, EVs, in turn, have become potential candidates as therapeutic agents. In this review, we discuss the evolving understanding of the role of EVs in CVD, summarize the current knowledge of EV-mediated regulatory mechanisms, and highlight potential strategies for the diagnosis and therapy of CVD. We also attempt to look into the future that may advance our understanding of the role of EVs in the pathogenesis of CVD and provide novel insights into the field of translational medicine.
Collapse
Affiliation(s)
- Jian Yang
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.
| | - Xue Zou
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, PR China; Chongqing Institute of Cardiology and Chongqing Key Laboratory for Hypertension Research, Chongqing, PR China
| | - Pedro A Jose
- Division of Renal Disease & Hypertension, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, PR China; Chongqing Institute of Cardiology and Chongqing Key Laboratory for Hypertension Research, Chongqing, PR China; State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, The Third Military Medical University, Chongqing, PR China; Heart Center of Fujian Province, Union Hospital, Fujian Medical University, Fuzhou, PR China.
| |
Collapse
|
14
|
Badimon L, Suades R, Vilella-Figuerola A, Crespo J, Vilahur G, Escate R, Padro T, Chiva-Blanch G. Liquid Biopsies: Microvesicles in Cardiovascular Disease. Antioxid Redox Signal 2020; 33:645-662. [PMID: 31696726 DOI: 10.1089/ars.2019.7922] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Significance: Circulating microvesicles (cMV) are small (0.1-1 μm) phospholipid-rich blebs released by almost all cell types, and their release increases with cell activation and injury, thus reflecting the state of the cell from which they are originated. Microvesicles (MV) are found in the bloodstream, and they affect the phenotype of recipient cells, after local or systemic circulation, by intercellular transfer of their molecular content. Recent Advances: Several studies suggest the use of cell-specific MV subpopulations as predictive biomarkers for cardiovascular diseases (CVDs) at different stages and degrees of severity. In this review, we describe the state of the art of cMV as noninvasive surrogate biomarkers of vascular injury and dysfunction correlated with poor clinical outcomes in CVD. Critical Issues: Despite the growing body of evidence supporting the importance of cMV as hallmarks of CVD and their utility as biomarkers of CVD, the specific roles of each phenotype of cMV in CVD burden and prognosis still remain to be elucidated and validated in large cohorts. In addition, the development of standardized and reproducible techniques is required to be used as biomarkers for disease progression in the clinical setting. Future Directions: A multipanel approach with specific cMV phenotypes, added to current biomarkers and scores, will undoubtedly provide unique prognostic information to stratify patients for appropriate therapy on the basis of their risk of atherothrombotic disease and will open a new research area as therapeutic targets for CVD. MV will add to the implementation of precision medicine by helping the cellular and molecular characterization of CVD patients.
Collapse
Affiliation(s)
- Lina Badimon
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Rosa Suades
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain.,Cardiology Unit, Department of Medicine Solna, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Alba Vilella-Figuerola
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain
| | - Javier Crespo
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain
| | - Gemma Vilahur
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Rafael Escate
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Teresa Padro
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Gemma Chiva-Blanch
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain
| |
Collapse
|
15
|
Extracellular vesicles in atrial fibrillation and stroke. Thromb Res 2020; 193:180-189. [PMID: 32717643 DOI: 10.1016/j.thromres.2020.07.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/08/2020] [Accepted: 07/10/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is associated with a 5-fold increased risk of thromboembolic stroke. Extracellular vesicles (EVs) convey pathophysiological information and are possible biomarkers for risk of stroke. METHODS EVs were measured in 836 patients with AF (of which 280 were stroke cases) selected from the ARISTOTLE trial and in a cohort of unselected 70 year old individuals (n = 1007, reference material). EVs from platelets, leukocytes, erythrocytes and inflammatory endothelial cells were measured using flow cytometry and a solid-phase proximity ligation assay. RESULTS Concentrations of EVs were higher in the ARISTOTLE patients than in the PIVUS cohort for all the EV groups except EVs from endothelial cells (p < 0.0001). The distributions of the concentrations of the EVs were similar among the control group and the stroke cases for all of the sources of EVs in the ARISTOTLE study. EVs were modestly correlated with the levels of NT-ProBNP, Cystatin C, GDF-15 and D-dimer. Stronger correlations were found for platelet EVs as well as phosphatidyl serine positive EVs that were correlated with CD40 ligand in the ARISTOTLE study. Leukocyte EVs were correlated with IL-6 in both the ARISTOTLE and the PIVUS study, implicating them in different physiological processes. CONCLUSIONS Higher levels of EVs were found in anticoagulated patients with AF and a higher risk of stroke than in a general population of similar age, possibly due to the high disease burden in AF patients. Our data with EVs representing a broad repertoire of activated blood cells in AF patients suggest that EVs are likely not a key mediator of occurrence of stroke in this population.
Collapse
|
16
|
Wang H, Song NP, Li JP, Wang ZH, Ti Y, Li YH, Zhang W, Zhong M. The microvesicle/CD36 complex triggers a prothrombotic phenotype in patients with non-valvular atrial fibrillation. J Cell Mol Med 2020; 24:7331-7340. [PMID: 32510852 PMCID: PMC7339157 DOI: 10.1111/jcmm.15311] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 01/15/2020] [Accepted: 03/28/2020] [Indexed: 02/02/2023] Open
Abstract
The mechanisms responsible for platelet activation, the prothrombotic state, in non-valvular atrial fibrillation (NVAF) are still obscure. Microvesicles (MVs) can transfer various messages to target cells and may be helpful for exploring the detailed mechanisms. We aimed to investigate the possible mechanisms by which proatherogenic factors of NVAF contribute to platelet activation. Two hundred and ten patients with NVAF were stratified as being at 'low to moderate risk' or 'high risk' for stroke according to the CHADS2 score. Levels of platelet-derived MVs (PMVs) and platelet activation were examined. CD36-positive or CD36-deficient human platelets were stimulated by MVs isolated from NVAF patients with or without various inhibitors in vitro. Levels of PMVs and platelet activation markers enhanced significantly in high-risk patients. The MVs isolated from plasma of NVAF patients bound to platelet CD36 and activated platelets by phosphorylating the mitogen-activated protein kinase 4/Jun N-terminal kinase 2 (MKK4/JNK2) pathways. However, CD36 deficiency protected against MV-induced activation of platelets. We reveal a possible mechanism of platelet activation in NVAF and suggest that the platelet CD36 might be an effective target in preventing the prothrombotic state in NVAF.
Collapse
Affiliation(s)
- Hua Wang
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao Medical College, Qingdao University, Yantai, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Nian-Peng Song
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao Medical College, Qingdao University, Yantai, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jian-Ping Li
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao Medical College, Qingdao University, Yantai, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Zhi-Hao Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.,Department of Geriatric Medicine, Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Yun Ti
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yi-Hui Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.,Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Wei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Ming Zhong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
17
|
Hasan H, Park SH, Auger C, Belcastro E, Matsushita K, Marchandot B, Lee HH, Qureshi AW, Kauffenstein G, Ohlmann P, Schini-Kerth VB, Jesel L, Morel O. Thrombin Induces Angiotensin II-Mediated Senescence in Atrial Endothelial Cells: Impact on Pro-Remodeling Patterns. J Clin Med 2019; 8:jcm8101570. [PMID: 31581517 PMCID: PMC6833093 DOI: 10.3390/jcm8101570] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 09/16/2019] [Accepted: 09/25/2019] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Besides its well-known functions in hemostasis, thrombin plays a role in various non-hemostatic biological and pathophysiologic processes. We examined the potential of thrombin to promote premature atrial endothelial cells (ECs) senescence. METHODS AND RESULTS Primary ECs were isolated from porcine atrial tissue. Endothelial senescence was assessed by measuring beta-galactosidase (SA-β-gal) activity using flow cytometry, oxidative stress using the redox-sensitive probe dihydroethidium, protein level by Western blot, and matrix metalloproteinases (MMPs) activity using zymography. Atrial endothelial senescence was induced by thrombin at clinically relevant concentrations. Thrombin induced the up-regulation of p53, a key regulator in cellular senescence and of p21 and p16, two cyclin-dependent kinase inhibitors. Nicotinamide adenine dinucleotide phosphate NADPH oxidase, cyclooxygenases and the mitochondrial respiration complex contributed to oxidative stress and senescence. Enhanced expression levels of vascular cell adhesion molecule (VCAM)-1, tissue factor, transforming growth factor (TGF)-β and MMP-2 and 9 characterized the senescence-associated secretory phenotype of atrial ECs. In addition, the pro-senescence endothelial response to thrombin was associated with an overexpression of both angiotensin converting enzyme and AT1 receptors and was inhibited by perindoprilat and losartan. CONCLUSIONS Thrombin promotes premature ageing and senescence of atrial ECs and may pave the way to deleterious remodeling of atrial tissue by a local up-regulation of the angiotensin system and by promoting pro-inflammatory, pro-thrombotic, pro-fibrotic and pro-remodeling responses. Hence, targeting thrombin and/or angiotensin systems may efficiently prevent atrial endothelial senescence.
Collapse
Affiliation(s)
- Hira Hasan
- INSERM UMR1260 Regenerative NanoMedicine, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Faculté de Pharmacie, BP 60024 FR-67401 Strasbourg, France
| | - Sin-Hee Park
- INSERM UMR1260 Regenerative NanoMedicine, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Faculté de Pharmacie, BP 60024 FR-67401 Strasbourg, France
| | - Cyril Auger
- INSERM UMR1260 Regenerative NanoMedicine, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Faculté de Pharmacie, BP 60024 FR-67401 Strasbourg, France
| | - Eugenia Belcastro
- INSERM UMR1260 Regenerative NanoMedicine, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Faculté de Pharmacie, BP 60024 FR-67401 Strasbourg, France
| | - Kensuke Matsushita
- INSERM UMR1260 Regenerative NanoMedicine, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Faculté de Pharmacie, BP 60024 FR-67401 Strasbourg, France
| | - Benjamin Marchandot
- Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, BP 426-67091 France
| | - Hyun-Ho Lee
- INSERM UMR1260 Regenerative NanoMedicine, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Faculté de Pharmacie, BP 60024 FR-67401 Strasbourg, France
| | - Abdul Wahid Qureshi
- INSERM UMR1260 Regenerative NanoMedicine, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Faculté de Pharmacie, BP 60024 FR-67401 Strasbourg, France
| | - Gilles Kauffenstein
- INSERM UMR1260 Regenerative NanoMedicine, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Faculté de Pharmacie, BP 60024 FR-67401 Strasbourg, France.
| | - Patrick Ohlmann
- Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, BP 426-67091 France
| | - Valérie B Schini-Kerth
- INSERM UMR1260 Regenerative NanoMedicine, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Faculté de Pharmacie, BP 60024 FR-67401 Strasbourg, France
| | - Laurence Jesel
- INSERM UMR1260 Regenerative NanoMedicine, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Faculté de Pharmacie, BP 60024 FR-67401 Strasbourg, France
- Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, BP 426-67091 France
| | - Olivier Morel
- INSERM UMR1260 Regenerative NanoMedicine, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Faculté de Pharmacie, BP 60024 FR-67401 Strasbourg, France.
- Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, BP 426-67091 France.
| |
Collapse
|
18
|
Dong X, Li M, Li Q, Gao Y, Liu L, Chen X, Zhou Z, Rong H, Zhang J, Tian Y. Effects of Cryopreservation on Microparticles Concentration, Procoagulant Function, Size Distribution, and Morphology. Med Sci Monit 2019; 25:6675-6690. [PMID: 31488807 PMCID: PMC6752109 DOI: 10.12659/msm.917962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background Research on microparticles is rapidly evolving and has extended to the field of many diseases. It is unclear whether microparticles can be stored frozen. In this study, our goal was to verify whether cryopreservation had an effect on the properties of the microparticles. Material/Methods We obtained C57BL/6J mouse-derived microparticles by grinding and gradient centrifugation. The specimens were divided into 2 groups: without dimethyl sulfoxide and with dimethyl sulfoxide. The microparticles were then stored at 25°C, 4°C, −20°C, −80°C, and −196°C for 0.5 days, 1 day, 3 days, 5 days, and 7 days. We tested whether the concentration, coagulation function, diameter distribution, and morphology of the microparticles in the 2 groups changed compared to those of a fresh sample. Results We discovered that the concentrations of total microparticles, annexin V-positive microparticles, and brain-derived microparticles changed with freezing. The coagulation function, morphology, and size distribution of the microparticles were also affected by cryopreservation. Finally, there was no difference in the effects of cryopreservation on microparticles between the dimethyl sulfoxide group and the dimethyl sulfoxide-free group. Conclusions This study suggests that cryopreservation has diverse effects on microparticles within 1 week and that dimethyl sulfoxide has no protective effect on cryopreserved microparticles. Therefore, microparticles should be used fresh for future studies, and they should not be cryopreserved with or without dimethyl sulfoxide.
Collapse
Affiliation(s)
- Xinlong Dong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education in China and Tianjin, Tianjin, China (mainland)
| | - Mengqi Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education in China and Tianjin, Tianjin, China (mainland)
| | - Qifeng Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education in China and Tianjin, Tianjin, China (mainland)
| | - Yalong Gao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education in China and Tianjin, Tianjin, China (mainland)
| | - Li Liu
- Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education in China and Tianjin, Tianjin, China (mainland)
| | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education in China and Tianjin, Tianjin, China (mainland)
| | - Ziwei Zhou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education in China and Tianjin, Tianjin, China (mainland)
| | - Hongtao Rong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education in China and Tianjin, Tianjin, China (mainland)
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education in China and Tianjin, Tianjin, China (mainland)
| | - Ye Tian
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education in China and Tianjin, Tianjin, China (mainland)
| |
Collapse
|
19
|
Abstract
Microparticles are a distinctive group of small vesicles, without nucleus, which are involved as significant modulators in several physiological and pathophysiological mechanisms. Plasma microparticles from various cellular lines have been subject of research. Data suggest that they are key players in development and manifestation of cardiovascular diseases and their presence, in high levels, is associated with chronic inflammation, endothelial damage and thrombosis. The strong correlation of microparticle levels with several outcomes in cardiovascular diseases has led to their utilization as biomarkers. Despite the limited clinical application at present, their significance emerges, mainly because their detection and enumeration methods are improving. This review article summarizes the evidence derived from research, related with the genesis and the function of microparticles in the presence of various cardiovascular risk factors and conditions. The current data provide a substrate for several theories of how microparticles influence various cellular mechanisms by transferring biological information.
Collapse
Affiliation(s)
- Christos Voukalis
- a Institute of Cardiovascular Sciences , University of Birmingham , Birmingham , UK
| | - Eduard Shantsila
- a Institute of Cardiovascular Sciences , University of Birmingham , Birmingham , UK
| | - Gregory Y H Lip
- b Liverpool Centre for Cardiovascular Science , University of Liverpool and Liverpool Heart & Chest Hospital , Liverpool , UK.,c Department of Clinical Medicine, Aalborg Thrombosis Research Unit , Aalborg University , Aalborg , Denmark
| |
Collapse
|
20
|
Chu G, Versteeg HH, Verschoor AJ, Trines SA, Hemels MEW, Ay C, Huisman MV, Klok FA. Atrial fibrillation and cancer - An unexplored field in cardiovascular oncology. Blood Rev 2019; 35:59-67. [PMID: 30928168 DOI: 10.1016/j.blre.2019.03.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/15/2019] [Accepted: 03/22/2019] [Indexed: 12/28/2022]
Abstract
An increasing body of evidence suggests an association between cancer and atrial fibrillation (AF). The exact magnitude and underlying mechanism of this association are however unclear. Cancer-related inflammation, anti-cancer treatment and other cancer-related comorbidities are proposed to affect atrial remodelling, increasing the susceptibility of cancer patients for developing AF. Moreover, cancer is assumed to modify the risk of thromboembolisms and bleeding. A thorough and adequate understanding of these risks is however lacking, as current literature is scarce and show ambiguous results in AF patients. The standardized risk-models that normally aid the clinician in the decision of initiating anticoagulant therapy do not take the presence of malignancy into account. Other factors that complicate risk assessment in AF patients with cancer include drug-drug interactions and other cancer-related comorbidities such as renal impairment. In this review, we highlight the available literature regarding epidemiological association, risk assessment and anticoagulation therapy in AF patients with cancer.
Collapse
Affiliation(s)
- Gordon Chu
- Department of Thrombosis and Haemostasis, Leiden University Medical Centre, the Netherlands.
| | - Henri H Versteeg
- Department of Thrombosis and Haemostasis, Leiden University Medical Centre, the Netherlands
| | - Arie J Verschoor
- Department of Medical Oncology, Leiden University Medical Centre, the Netherlands
| | - Serge A Trines
- Department of Cardiology, Heart-Lung Centre, Leiden University Medical Centre, the Netherlands
| | - Martin E W Hemels
- Department of Cardiology, Rijnstate, Arnhem, the Netherlands; Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cihan Ay
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Austria
| | - Menno V Huisman
- Department of Thrombosis and Haemostasis, Leiden University Medical Centre, the Netherlands
| | - Frederikus A Klok
- Department of Thrombosis and Haemostasis, Leiden University Medical Centre, the Netherlands
| |
Collapse
|
21
|
Morphological determinators of platelet activation status in patients with atrial fibrillation. Int J Cardiol 2019; 279:90-95. [DOI: 10.1016/j.ijcard.2018.11.096] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 11/04/2018] [Accepted: 11/15/2018] [Indexed: 01/12/2023]
|
22
|
Fender AC. Connecting atrial morphology with platelet reactivity: What the left atrial appendage can tell us about the procoagulant state in atrial fibrillation. Int J Cardiol 2019; 279:112-113. [DOI: 10.1016/j.ijcard.2018.12.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 12/18/2018] [Indexed: 11/30/2022]
|
23
|
Siwaponanan P, Keawvichit R, Udompunturak S, Hunnangkul S, Reesukumal K, Sukapirom K, Pattanapanyasat K, Krittayaphong R. Altered profile of circulating microparticles in nonvalvular atrial fibrillation. Clin Cardiol 2019; 42:425-431. [PMID: 30680757 PMCID: PMC6712324 DOI: 10.1002/clc.23158] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/16/2019] [Accepted: 01/19/2019] [Indexed: 12/12/2022] Open
Abstract
Background Nonvalvular atrial fibrillation (AF) is the most common cardiac arrhythmia, and it is associated with the prothrombotic state. Circulating microparticles (cMPs) are membrane vesicles that are shed from many cell types in response to cell activation and cell apoptosis. Several studies reported that cMPs may play a role in the hypercoagulable state that can be observed in patients with AF. The aim of this study was to determine the levels of total cMPs and characterize their cellular origins in AF patients. Methods Atotal of 66 AF patients and 33 healthy controls were enrolled. This study investigated total cMP levels and their cellular origin in AF patients using polychromatic flow cytometry. Results AF patients had significantly higher levels of total cMPs (median 36.38, interquartile range [IQR] 21.16‐68.50 × 105 counts/mL vs median 15.21, IQR 9.91‐30.86 × 105 counts/mL; P = 0.004), platelet‐derived MPs (PMPs) (median 10.61, IQR 6.55‐18.04 × 105 counts/mL vs median 7.83, IQR 4.44‐10.26 × 10/mL; P = 0.009), and endothelial‐derived MPs (EMPs CD31+ CD41−) (median 2.94, IQR 1.78‐0.60 × 105 counts/mL vs median 1.16, IQR 0.71‐2.30 × 105 counts/mL; P = 0.001) than healthy controls after adjusting for potential confounders. Phosphatidylserine positive MP (PS + MP) levels were similar compared between AF patients and healthy controls. Conclusion The results of this study revealed a marked increase in total cMP levels, and evidence of elevated endothelial damage and platelet activation, as demonstrated by increased PMP and EMP levels, in AF patients. Additional study is needed to further elucidate the role of cMPs (PMPs and EMPs) in the pathophysiology of and the complications associated with AF.
Collapse
Affiliation(s)
- Panjaree Siwaponanan
- Division of Cardiology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Siriraj Center of Research Excellence in Microparticle and Exosome in Diseases, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Rassamon Keawvichit
- Siriraj Center of Research Excellence in Microparticle and Exosome in Diseases, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Suthipol Udompunturak
- Division of Clinical Epidemiology, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Saowalak Hunnangkul
- Division of Clinical Epidemiology, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kanit Reesukumal
- Department of Clinical Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kasama Sukapirom
- Siriraj Center of Research Excellence in Microparticle and Exosome in Diseases, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kovit Pattanapanyasat
- Siriraj Center of Research Excellence in Microparticle and Exosome in Diseases, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Rungroj Krittayaphong
- Division of Cardiology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
24
|
Elevated blood plasma levels of tissue factor-bearing extracellular vesicles in patients with atrial fibrillation. Thromb Res 2019; 173:141-150. [DOI: 10.1016/j.thromres.2018.11.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 11/05/2018] [Accepted: 11/27/2018] [Indexed: 02/06/2023]
|
25
|
Gohar A, de Kleijn DP, Hoes AW, Rutten FH, Hilfiker-Kleiner D, Ferdinandy P, Sluijter JP, den Ruijter HM. Vascular extracellular vesicles in comorbidities of heart failure with preserved ejection fraction in men and women: The hidden players. A mini review. Vascul Pharmacol 2018; 111:1-6. [DOI: 10.1016/j.vph.2018.05.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 03/07/2018] [Accepted: 05/18/2018] [Indexed: 12/13/2022]
|
26
|
Phosphatidylserine-exposing blood cells and microparticles induce procoagulant activity in non-valvular atrial fibrillation. Int J Cardiol 2018; 258:138-143. [PMID: 29544920 DOI: 10.1016/j.ijcard.2018.01.116] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/21/2018] [Accepted: 01/26/2018] [Indexed: 11/23/2022]
Abstract
BACKGROUND The definitive role of phosphatidylserine (PS) in the prothrombotic state of non-valvular atrial fibrillation (NVAF) remains unclear. Our objectives were to study the PS exposure on blood cells and microparticles (MPs) in NVAF, and evaluate their procoagulant activity (PCA). METHODS NVAF patients without (n = 60) and with left atrial thrombi (n = 18) and controls (n = 36) were included in our study. Exposed PS was analyzed with flow cytometry and confocal microscopy. PCA was evaluated using clotting time, factor Xa (FXa), thrombin and fibrin formation. RESULTS PS+ blood cells and MPs were significantly higher in NVAF patients without and with left atrial thrombi (both P < 0.01) than in controls. Patients with left atrial thrombi showed increased PS+ platelets, neutrophils, erythrocytes and MPs compared with patients without thrombi (all P < 0.05). Moreover, in patients with left atrial thrombi, MPs primarily originated from platelets (56.1%) followed by leukocytes (21.9%, including MPs from neutrophils, monocytes and lymphocytes), erythrocytes (12.2%) and endothelial cells (8.9%). Additionally, PS+ blood cells and MPs contributed to markedly shortened coagulation time and dramatically increased FXa/thrombin/fibrin (all P < 0.001) generation in both NVAF groups. Furthermore, blockade of exposed PS on blood cells and MPs with lactadherin inhibited PCA by approximately 80%. Lastly, we found that the amount of PS+ platelets and MPs was positively correlated with thrombus diameter (all p < 0.005). CONCLUSIONS Our results suggest that exposed PS on blood cells and MPs play a procoagulant role in NVAF patients. Blockade of PS prior to thrombus formation might be a novel therapeutic approach in these patients.
Collapse
|
27
|
Pourtau L, Sellal JM, Lacroix R, Poncelet P, Bernus O, Clofent-Sanchez G, Hocini M, Haïssaguerre M, Dignat-George F, Sacher F, Nurden P. Platelet function and microparticle levels in atrial fibrillation: Changes during the acute episode. Int J Cardiol 2018; 243:216-222. [PMID: 28747025 DOI: 10.1016/j.ijcard.2017.03.068] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 03/08/2017] [Accepted: 03/10/2017] [Indexed: 11/24/2022]
Abstract
BACKGROUND Thrombotic risk constitutes a major complication of atrial fibrillation (AF). Platelets and microparticles (MPs) are important for hemostasis and thrombosis, however their participation during AF is not well known. The aim of this study was to characterize platelet function and MPs procoagulant and fibrinolytic activity in AF patients and to determine the effects of an acute-AF episode. METHODS Blood was collected from paroxysmal (21) and persistent (16) AF patients referred for AF catheter ablation. Ten patients in sinus rhythm for 10days were induced in AF allowing comparisons of left atrium samples before and after induction. Platelet aggregation with ADP, TRAP, collagen, and ristocetin was studied. Platelet surface expression of PAR-1, αIIbβ3, GPIb and P-selectin were evaluated by flow cytometry, and MPs-associated procoagulant and fibrinolytic activity levels were determined by functional assays. RESULTS A specific reduction in platelet aggregation to TRAP, activating the thrombin receptor PAR-1, was found in all AF patients. No differences in platelet receptor expression were found. Yet, after acute-induced AF, the platelet response was improved. Furthermore, a significant decrease of left atrium tissue factor-dependent procoagulant activity of MPs was observed. CONCLUSION Acute episodes of AF results in a decrease in MPs-associated tissue factor activity, possibly corresponding to consumption, which in turn favors coagulation and the local production of thrombin. A decreased platelet basal aggregation to TRAP may result from PAR1 desensitization, whereas the improved response after an induced episode of AF suggests activation of coagulation and PAR1 re-sensitization.
Collapse
Affiliation(s)
- Line Pourtau
- IHU Liryc, Electrophysiology and Heart Modeling Institute, fondation Bordeaux Université, 33600, Pessac, France; Univ. Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, 33000 Bordeaux, France; INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, 33000 Bordeaux, France.
| | - Jean Marc Sellal
- IHU Liryc, Electrophysiology and Heart Modeling Institute, fondation Bordeaux Université, 33600, Pessac, France; Bordeaux University Hospital (CHU), Electrophysiology and Ablation Unit, 33600 Pessac, France; Centre Hospitalier Régional Universitaire (CHRU) de Nancy, département de cardiologie, 54500 Vandœuvre-lès-Nancy, France.
| | - Romaric Lacroix
- VRCM, UMR-S1076, Aix -Marseille Université, INSERM, UFR de Pharmacie, 13385 Marseille, France; Department of Haematology and Vascular Biology, CHU Conception, AP-HM, 13385 Marseille, France.
| | - Philippe Poncelet
- Research & Technology Department, BioCytex, 13010 Marseille, France.
| | - Olivier Bernus
- IHU Liryc, Electrophysiology and Heart Modeling Institute, fondation Bordeaux Université, 33600, Pessac, France; Univ. Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, 33000 Bordeaux, France; INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, 33000 Bordeaux, France.
| | - Gisèle Clofent-Sanchez
- Univ Bordeaux, CNRS, Centre de Résonance Magnétique des Systèmes Biologiques, U5536, 33076 Bordeaux, France.
| | - Mélèze Hocini
- IHU Liryc, Electrophysiology and Heart Modeling Institute, fondation Bordeaux Université, 33600, Pessac, France; Univ. Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, 33000 Bordeaux, France; INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, 33000 Bordeaux, France; Bordeaux University Hospital (CHU), Electrophysiology and Ablation Unit, 33600 Pessac, France.
| | - Michel Haïssaguerre
- IHU Liryc, Electrophysiology and Heart Modeling Institute, fondation Bordeaux Université, 33600, Pessac, France; Univ. Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, 33000 Bordeaux, France; INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, 33000 Bordeaux, France; Bordeaux University Hospital (CHU), Electrophysiology and Ablation Unit, 33600 Pessac, France.
| | - Françoise Dignat-George
- VRCM, UMR-S1076, Aix -Marseille Université, INSERM, UFR de Pharmacie, 13385 Marseille, France; Department of Haematology and Vascular Biology, CHU Conception, AP-HM, 13385 Marseille, France.
| | - Frédéric Sacher
- IHU Liryc, Electrophysiology and Heart Modeling Institute, fondation Bordeaux Université, 33600, Pessac, France; Univ. Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, 33000 Bordeaux, France; INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, 33000 Bordeaux, France; Bordeaux University Hospital (CHU), Electrophysiology and Ablation Unit, 33600 Pessac, France.
| | - Paquita Nurden
- IHU Liryc, Electrophysiology and Heart Modeling Institute, fondation Bordeaux Université, 33600, Pessac, France.
| |
Collapse
|
28
|
Reiffel JA. Optimum Risk Assessment for Stroke in Atrial Fibrillation: Should We Hold the Status Quo or Consider Magnitude Synergism and Left Atrial Appendage Anatomy? Arrhythm Electrophysiol Rev 2017; 6:161-166. [PMID: 29326830 PMCID: PMC5739887 DOI: 10.15420/aer.2017.33.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 10/09/2017] [Indexed: 01/06/2023] Open
Abstract
Thromboembolic stroke and systemic embolism are generally agreed to be the major morbidity/mortality concerns for patients with AF. However, the risk of thromboembolism is not the same for all AF patients. Both AF and comorbidities must interact synergistically to create the risk for thromboembolism. But, is the synergism dichotomous - AF present or absent, comorbid disorder present or absent - or does synergism have magnitude, depending on the number and severity of the associated disorders and the amount of time one is in AF? This review discusses the current risk-score contributors and options for assessing risk of thromboembolism in AF patients, and what their combined roles might be. Also covered is the consideration of left atrial appendage anatomy in this context.
Collapse
|
29
|
Potential Biological Markers of Atrial Fibrillation: A Chance to Prevent Cryptogenic Stroke. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8153024. [PMID: 28785588 PMCID: PMC5530434 DOI: 10.1155/2017/8153024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/15/2017] [Accepted: 06/04/2017] [Indexed: 01/04/2023]
Abstract
Stroke affects millions of people all over the world, causing death and disability. The most frequent type of this disease is ischemic stroke, which can be caused by different factors. In approximately 25 percent of cases, no obvious cause can be found. Recent observations have shown that paroxysmal atrial fibrillation could be responsible for a significant number of cryptogenic stroke events. Short- or long-lasting ECG monitoring could help with the diagnosis of transient arrhythmias. Unfortunately, these techniques either are expensive or require good patient compliance. An alternative option is the identification of biological markers that are specific for atrial fibrillation and can be used to predict arrhythmia. In this review, we give a summary of the recent advances in the research of arrhythmia markers. Based on their structure and function, we differentiated four groups of biomarkers: markers of inflammation, markers of fibrosis, markers with hormonal activity, and other markers. In spite of intensive researches, the optimal biological marker is still not available, but there are some promising markers, like NT-proBNP/BNP.
Collapse
|
30
|
Lau YC, Xiong Q, Blann AD, Lip GYH. Relationship between renal function and circulating microparticles, soluble P-selectin and E-selectin levels in atrial fibrillation. J Thromb Thrombolysis 2017; 43:18-23. [PMID: 27671694 PMCID: PMC5233739 DOI: 10.1007/s11239-016-1427-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Atrial fibrillation (AF) and chronic kidney disease are closely related, and any associated risk of stroke and thromboembolism due to AF is increased by concurrent renal dysfunction. The mechanism(s) for this include abnormalities in platelets and endothelial cells. We hypothesized relationships between levels of circulating platelet microparticles (PMPs, defined by CD42b), soluble P selectin (both reflecting platelet activation), soluble E-selectin (reflecting endothelial activation) and endothelial/platelet microparticles (EPMPs, defined by CD31) with progressive renal dysfunction. Blood samples were obtained from 160 anticoagulated AF patients. Microparticles were measured by flow cytometry, soluble E and P selectin levels by ELISA. Renal function was determined by estimated glomerular filtration rate (eGFR). EPMP levels demonstrated a linear increased trend across quartiles of eGFR (p = 0.034) and CKD stage (p < 0.001), and correlated with eGFR and serum creatinine (p < 0.01). PMPs, P-selectin and E-selectin levels were not significantly different across groupings of renal dysfunction, and no significant correlations with eGFR were evident (p = 0.186, p = 0.561, p = 0.746 respectively). Stepwise multivariable regression analysis demonstrated that worsening renal function was an independent predictor of EPMP levels (p < 0.001). In well-anticoagulated AF patients, there is potential relationship between endothelial function (as judged by elevated EPMP levels, with no change in PMPs) and renal function. Other markers of prothombotic state or cellular activation (PMP, P-selectin and E-selectin levels) were not significantly different across the various degree of renal dysfunction. Renal function must be addressed when measuring EPMP levels.
Collapse
Affiliation(s)
- Yee Cheng Lau
- University of Birmingham Institute of Cardiovascular Science, City Hospital, Dudley Road, Birmingham, UK
| | - Qinmei Xiong
- University of Birmingham Institute of Cardiovascular Science, City Hospital, Dudley Road, Birmingham, UK.,Cardiovascular Department, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Andrew D Blann
- University of Birmingham Institute of Cardiovascular Science, City Hospital, Dudley Road, Birmingham, UK
| | - Gregory Y H Lip
- University of Birmingham Institute of Cardiovascular Science, City Hospital, Dudley Road, Birmingham, UK.
| |
Collapse
|
31
|
Extracellular vesicles and blood diseases. Int J Hematol 2017; 105:392-405. [PMID: 28130731 DOI: 10.1007/s12185-017-2180-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/09/2017] [Accepted: 01/09/2017] [Indexed: 01/19/2023]
Abstract
Extracellular vesicles (EVs) are small membrane vesicles released from many different cell types by the exocytic budding of the plasma membrane in response to cellular activation or apoptosis. EVs disseminate various bioactive effectors originating from the parent cells and transfer functional RNA and protein between cells, enabling them to alter vascular function and induce biological responses involved in vascular homeostasis. Although most EVs in human blood originate from platelets, EVs are also released from leukocytes, erythrocytes, endothelial cells, smooth muscle cells, and cancer cells. EVs were initially thought to be small particles with procoagulant activity; however, they can also evoke cellular responses in the immediate microenvironments and transport microRNAs (miRNA) into target cells. In this review, we summarize the recent literature relevant to EVs, including a growing list of clinical disorders that are associated with elevated EV levels. These studies suggest that EVs play roles in various blood diseases.
Collapse
|
32
|
Thulin Å, Christersson C, Alfredsson J, Siegbahn A. Circulating cell-derived microparticles as biomarkers in cardiovascular disease. Biomark Med 2016; 10:1009-22. [PMID: 27586235 DOI: 10.2217/bmm-2016-0035] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular diseases (CVDs) are a common cause of death, and a search for biomarkers for risk stratification is warranted. Elevated levels of cell-derived microparticles (MPs) are found in patients with CVD and in groups with risk factors for CVD. Subpopulations of MPs are promising biomarkers for improving risk prediction, as well as monitoring treatment. However, the field has been hampered by technical difficulties, and the ongoing development of sensitive standardized techniques is crucial for implementing MP analyses in the clinic. Large prospective studies are required to establish which MPs are of prognostic value in different patient groups. In this review, we discuss methodological challenges and progress in the field, as well as MP populations that are of interest for further clinical evaluation.
Collapse
Affiliation(s)
- Åsa Thulin
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Sweden
| | | | - Jenny Alfredsson
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Sweden
| | - Agneta Siegbahn
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Sweden
| |
Collapse
|
33
|
Amiral J, Seghatchian J. Measurement of extracellular vesicles as biomarkers of consequences or cause complications of pathological states, and prognosis of both evolution and therapeutic safety/efficacy. Transfus Apher Sci 2016; 55:23-34. [PMID: 27475803 DOI: 10.1016/j.transci.2016.07.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Utility of EVs, as biomarkers of cause or consequence of various pathological complications, and prognosis of blood components' therapy in terms of safety/efficacy and their potential associated hazards, primed by EVs involvements in pro-inflammatory, immunomodulatory and activations of both pro/anti-coagulatory and others associated pathways, as well as various cellular cross talks, are highlighted as the fundamental. Today EVs are becoming the "buzz" words of the current diagnosis, development and research [DDR] strategies, with the aim of ensuring safer therapeutic approaches in the current clinical practices, also incorporating their potential in long term cost effectiveness in health care systems. The main focus of this manuscript is to review the current opinions in some fundamental areas of EVs involvements in health and diseases. Firstly, our goal is highlighting what are EVs/MVs/MPs and how are they generated in physiology, pathology or blood products; classification and significance of EVs generated in vivo; followed by consequences and physiological/pathological induced effects of EVs generation in vivo. Secondly, specific cell origin EVs and association with malignancy; focus on EVs carrying TF and annexin V as a protective protein for harmful effects of EVs, and associations with LA; and incidence of anti-annexin V antibodies are also discussed. Thirdly, utility of EVs is presented: as diagnostic tools of disease markers; prognosis and follow-up of clinical states; evaluation of therapy efficacy; quality and risk assessment of blood products; followed by the laboratory tools for exploring, characterizing and measuring EVs, and/or their associated activity, using our own experiences of capture based assays. Finally, in perspective, the upcoming low volume sampling, fast, reliable and reproducibility and friendly use laboratory tools and the standardization of measurement methods are highlighted with the beneficial effects that we are witnessing in both wound healing and tissue remodeling, with an expected blockbuster status EVs as future therapeutic directions.
Collapse
Affiliation(s)
- Jean Amiral
- Hyphen BioMed, Neuville sur Oise, Paris, France.
| | - Jerard Seghatchian
- International Consultancy in Blood Components Quality/Safety Improvement, Audit/Inspection and DDR Strategies, London, UK
| |
Collapse
|
34
|
Ivak P, Pitha J, Netuka I. Circulating microparticles as a predictor of vascular properties in patients on mechanical circulatory support; hype or hope? Physiol Res 2016; 65:727-735. [PMID: 27429109 DOI: 10.33549/physiolres.933246] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Microparticles are small circulating vesicles originating from circulatory system and vascular wall cells released during their activation or damage. They possess different roles in regulation of endothelial function, inflammation, thrombosis, angiogenesis, and in general, cellular stress. Microparticles are the subject of intensive research in pulmonary hypertension, atherosclerotic disease, and heart failure. Another recently emerging role is the evaluation of the status of vasculature in end-stage heart failure patients treated with implantable ventricular assist devices. In patients implanted as destination therapy, assessment of the long-term effect of currently used continuous-flow left ventricular assist devices (LVADs) on vasculature might be of critical importance. However, unique continuous flow pattern generated by LVADs makes it difficult to assess reliably the vascular function with most currently used methods, based mainly on ultrasound detection of changes of arterial dilatation during pulsatile flow. In this respect, the measurement of circulating microparticles as a marker of vascular status may help to elucidate both short- and long-term effects of LVADs on the vascular system. Because data regarding this topic are very limited, this review is focused on the advantages and caveats of the circulating microparticles as markers of vascular function in patients on continuous-flow LVADs.
Collapse
Affiliation(s)
- P Ivak
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | | | | |
Collapse
|
35
|
Liles J, Liles J, Wanderling C, Syed M, Hoppensteadt D, Fareed J. Increased Level of Thrombotic Biomarkers in Patients with Atrial Fibrillation Despite Traditional and New Anticoagulant Therapy. Clin Appl Thromb Hemost 2016; 22:743-748. [PMID: 27170781 DOI: 10.1177/1076029616648407] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The aim of this study was to examine the effect of the traditional oral anticoagulant, warfarin (W), and new anticoagulants, apixaban (A) and rivaroxaban (R), on the level of thrombotic biomarkers in patients with atrial fibrillation (AF). Circulating plasma levels of von Willebrand factor (vWF), prothrombin fragment 1.2 (F1.2), microparticle tissue factor (MP-TF), and plasminogen activator inhibitor (PAI-1) were analyzed as potential markers of clot formation in 30 patients with AF prior to ablation surgery. Patients with AF were divided into 2 groups based on their usage (n = 21) and nonusage (n = 9) of any oral anticoagulant. Furthermore, those on anticoagulants were divided based on their use of newer (R and A, 16) or traditional (W, 4) anticoagulants. A statistical increase (P < .05) in the levels of vWF, MP-TF, and PAI-1 were seen in anticoagulated patients with AF, whereas F1.2 and PAI-1 were increased in nonanticoagulated patients with AF compared to normal. There was no statistical difference (P > .05) in levels of any thrombotic biomarker between patients treated with the traditional anticoagulant, W, and those treated with new anticoagulants, R and A. Our data suggest that, despite the use of traditional or newer anticoagulants, prothrombotic biomarkers are still generated at increased levels in patients with AF. Further studies to confirm these findings are warranted.
Collapse
Affiliation(s)
- Jeffrey Liles
- Stritch School of Medicine, Loyola University of Chicago, Maywood, IL, USA
| | - Jordan Liles
- Stritch School of Medicine, Loyola University of Chicago, Maywood, IL, USA
| | | | - Mushabbar Syed
- Department of Cardiology, Loyola University Medical Center, Maywood, IL, USA
| | - Debra Hoppensteadt
- Department of Pathology, Loyola University Medical Center, Maywood, IL, USA
| | - Jawed Fareed
- Department of Pathology, Loyola University Medical Center, Maywood, IL, USA
| |
Collapse
|
36
|
Berezin AE, Kremzer A, Berezina T, Martovitskaya Y. The signature of circulating microparticles in heart failure patients with metabolic syndrome. J Circ Biomark 2016; 5:1849454416663659. [PMID: 28936261 PMCID: PMC5548327 DOI: 10.1177/1849454416663659] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 07/15/2016] [Indexed: 11/15/2022] Open
Abstract
The role of pattern of circulating endothelial cell-derived microparticles, platelet-derived microparticles (PMPs), and monocyte-derived microparticles (MMPs) in metabolic syndrome (MetS) patients with chronic heart failure (CHF) is not still understood. The aim of the study was to investigate a pattern of circulating microparticles (MPs) in MetS patients with CHF in relation to neurohumoral and inflammatory activation. The study retrospectively involved 101 patients with MetS and 35 healthy volunteers. Biomarkers were measured at baseline of the study. The results of the study have shown that numerous circulating PMPs- and MMPs in subjects with MetS (with or without CHF) insufficiently distinguished from level obtained in healthy volunteers. We found elevated level of CD31+/annexin V+ MPs in association with lower level of CD62E+ MPs. Therefore, we found that biomarkers of biomechanical stress serum N-terminal brain natriuretic peptide and inflammation (high-sensitive C-reactive protein ,osteoprotegerin) remain statistically significant predictors for decreased CD62E+ to CD31+/annexin V+ ratio in MetS patients with CHF. In conclusion, decreased CD62E+ to CD31+/annexin V+ ratio reflected that impaired immune phenotype of MPs may be discussed as a surrogate marker of CHF development in MetS population.
Collapse
Affiliation(s)
- Alexander E Berezin
- Consultant of the Therapeutic Unit, Internal Medicine Department, State Medical University, Zaporozhye, Ukraine
| | - Alexander Kremzer
- Consultant of the Therapeutic Unit, Internal Medicine Department, State Medical University, Zaporozhye, Ukraine
| | | | | |
Collapse
|
37
|
Berezin AE, Kremzer AA, Berezina TA, Martovitskaya YV. Pattern of circulating microparticles in chronic heart failure patients with metabolic syndrome: Relevance to neurohumoral and inflammatory activation. BBA CLINICAL 2015; 4:69-75. [PMID: 26674662 PMCID: PMC4661711 DOI: 10.1016/j.bbacli.2015.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/21/2015] [Accepted: 07/31/2015] [Indexed: 01/15/2023]
Affiliation(s)
- Alexander E. Berezin
- Internal Medicine Department, State Medical University, 26, Mayakovsky av., Zaporozhye Postcode 69035, Ukraine
| | | | | | | |
Collapse
|
38
|
The diagnostic usefulness of capture assays for measuring global/specific extracellular micro-particles in plasma. Transfus Apher Sci 2015; 53:127-36. [PMID: 26572801 DOI: 10.1016/j.transci.2015.10.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Capture assays were developed and validated for measuring the global pro-coagulant activity of micro-particles (MPs, mainly originated from platelets), or specific extravascular cellular MPs (released from erythrocytes, leukocytes, monocytes, endothelial cells) as those exposing TF (MP-TF, mainly observed in patients with some cancers). Conversely to Flow Cytometry methods, these capture assays measure all coagulant activity associated with MPs, through thrombin generation (MP-Activity) or Factor Xa generation (MP-TF), and therefore they bring a complementary information, as they are more specific for the pro-coagulant activity associated with MPs. Small particles (<0.40 µ) exposing Phosphatidyl Serine (PS) exhibit a greater pro-coagulant surface than larger MPs (0.40 to >1.00 µ), those preferentially measured with flow cytometry. Activity associated with MPs is a consequence of disease but can also be a cause contributing to pathological processes and development of thrombo-embolic events. In many diseases, flow cytometry and capture assays do not totally correlate, and have different associations with disease evolution. Optimized capture based assays are presented and discussed, along with their performance characteristics and some applications. They can be performed in any technically skillful hemostasis laboratory, using a thermostated ELISA equipment, or an incubator. Dynamic ranges for MP-Activity assay is from <0.1 nM to >2.5 nM Phospholipids, expressed as Phosphatidyl Serine (PS) equivalent, in the tested dilution. For MP-TF the very sensitive bio-immunoassay reported allows measuring concentrations from <0.10 pg/ml (TF equivalent) to >5.00 pg/ml, in the assayed dilution. No measurable MP-TF was found in normals, although an important concentration was generated from whole blood treated with Lipo-Poly-Saccharides. Capture based assays are then highly useful in the laboratory setting for measuring the activities associated with pro-coagulant, or specific cellular MPs.
Collapse
|
39
|
Nomura S, Niki M, Nisizawa T, Tamaki T, Shimizu M. Microparticles as Biomarkers of Blood Coagulation in Cancer. BIOMARKERS IN CANCER 2015; 7:51-6. [PMID: 26462252 PMCID: PMC4592056 DOI: 10.4137/bic.s30347] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/05/2015] [Accepted: 08/07/2015] [Indexed: 12/20/2022]
Abstract
Cancer is associated with hypercoagulopathy and increased risk of thrombosis. This negatively influences patient morbidity and mortality. Cancer is also frequently complicated by the development of venous thromboembolism (VTE). Tumor-derived tissue factor (TF)-bearing microparticles (MPs) are associated with VTE events in malignancy. MPs are small membrane vesicles released from many different cell types by exocytic budding of the plasma membrane in response to cellular activation or apoptosis. MPs may also be involved in clinical diseases through expression of procoagulative phospholipids. The detection of TF-expressing MPs in cancer patients may be clinically useful. In lung and breast cancer patients, MPs induce metastasis and angiogenesis and may be indicators of vascular complications. Additionally, MPs in patients with various types of cancer possess adhesion proteins and bind target cells to promoting cancer progression or metastasis. Overexpression of TF by cancer cells is closely associated with tumor progression, and shedding of TF-expressing MPs by cancer cells correlates with the genetic status of cancer. Consequently, TF-expressing MPs represent important markers to consider in the prevention of and therapy for VTE complications in cancer patients.
Collapse
Affiliation(s)
- Shosaku Nomura
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Osaka, Japan
| | - Maiko Niki
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Osaka, Japan
| | - Tohru Nisizawa
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Osaka, Japan
| | - Takeshi Tamaki
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Osaka, Japan
| | - Michiomi Shimizu
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Osaka, Japan
| |
Collapse
|
40
|
Abstract
Microparticles (MPs) are small membrane vesicles that are released from many different cell types by exocytotic budding of the plasma membrane in response to cellular activation or apoptosis. MPs may be involved in both physiological processes and clinical treatments because they express phospholipids, which function as procoagulants. Elevated levels of platelet-derived MPs, endothelial cell-derived MPs, and monocyte-derived MPs are observed in almost all thrombotic diseases occurring in venous and arterial beds. Several studies have shown that the quantity, cellular origin, and composition of circulating MPs depend on the type of disease, the disease state, and medical treatment. Although MPs were initially thought to be small particles with only procoagulant activity, they are now known to have many different functions. An increasing number of studies have identified new implications of elevated MPs in clinical disorders. On the basis of evidence available till date, the present review suggests that MPs may be a useful biomarker in identifying atherothrombosis.
Collapse
Affiliation(s)
- Shosaku Nomura
- First Department of Internal Medicine, Kansai Medical University
| |
Collapse
|
41
|
Nomura S, Shimizu M. Clinical significance of procoagulant microparticles. J Intensive Care 2015; 3:2. [PMID: 25705427 PMCID: PMC4336124 DOI: 10.1186/s40560-014-0066-z] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 12/03/2014] [Indexed: 01/05/2023] Open
Abstract
Microparticles (MPs) are small membrane vesicles that are released from many different cell types by exocytic budding of the plasma membrane in response to cellular activation or apoptosis. MPs may also be involved in clinical diseases because they express phospholipids, which function as procoagulants. Although flow cytometry is the most widely used method for studying MPs, some novel assays, such as tissue factor-dependent procoagulant assay or the ELISA method, have been reported. However, the use of quantification of MP as a clinical tool is still controversial. Elevated platelet-derived MP, endothelial cell-derived MP, and monocyte-derived MP concentrations are documented in almost all thrombotic diseases occurring in venous and arterial beds. However, the significance of MPs in various clinical conditions remains controversial. An example of this controversy is that it is unknown if MPs found in peripheral blood vessels cause thrombosis or whether they are the result of thrombosis. Numerous studies have shown that not only the quantity, but also the cellular origin and composition of circulating MPs, are dependent on the type of disease, the disease state, and medical treatment. Additionally, many different functions have been attributed to MPs. Therefore, the number and type of clinical disorders associated with elevated MPs are currently increasing. However, MPs were initially thought to be small particles with procoagulant activity. Taken together, our review suggests that MPs may be a useful biomarker to identify thrombosis.
Collapse
Affiliation(s)
- Shosaku Nomura
- First Department of Internal Medicine, Kansai Medical University, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191 Japan
| | - Michiomi Shimizu
- First Department of Internal Medicine, Kansai Medical University, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191 Japan
| |
Collapse
|
42
|
Idriss NK, Blann AD, Sayed DM, Gaber MA, Hassen HA, Kishk YT. Circulating Endothelial Cells and Platelet Microparticles in Mitral Valve Disease With and Without Atrial Fibrillation. Angiology 2014; 66:631-7. [PMID: 25115553 DOI: 10.1177/0003319714546183] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Hypercoagulability in mitral valve disease (MVD), a cause of atrial fibrillation (AF) and stroke, is potentially due to endothelial damage/dysfunction (marked by circulating endothelial cells [CECs]), platelet activation (soluble P-selectin [sPsel], platelet microparticles [PMPs], and soluble CD40 [sCD40]), and oxidized low-density lipoprotein (oxLDL) cholesterol. We measured these variables in 24 patients with MVD as well as in 21 with MVD + AF and compared them with 20 healthy controls (HCs). The CECs and PMPs were measured by flow cytometry; sPsel, oxLDL, and CD40 by enzyme-linked immunosorbent assay. Compared with HCs, sPsel and PMPs were equally higher in MVD and MVD + AF; sCD40 and oxLDL were higher in MVD + AF than in HCs and MVD; and CECs were higher in MVD than in the HCs, with further increases in MVD + AF (all P < .001). We conclude that excess platelet activation is present in MVD regardless of AF, and that increased endothelial damage in MVD is greater when compounded by AF.
Collapse
Affiliation(s)
- Naglaa K Idriss
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Andrew D Blann
- Department of Medicine City Hospital, University of Birmingham Centre for Cardiovascular Sciences, Birmingham, United Kingdom
| | - Douaa M Sayed
- Department of Clinical Pathology, Faculty of Medicine, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Marwa A Gaber
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Hosny A Hassen
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Yehia Taha Kishk
- Department of Cardiology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
43
|
Freestone B, Krishnamoorthy S, Lip GYH. Assessment of endothelial dysfunction. Expert Rev Cardiovasc Ther 2014; 8:557-71. [DOI: 10.1586/erc.09.184] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
44
|
JESEL LAURENCE, ARENTZ THOMAS, HERRERA-SIKLODY CLAUDIA, TRENK DIETMAR, ZOBAIRI FATIHA, ABBAS MALAK, WEBER REINHOLD, MINNERS JAN, TOTI FLORENCE, MOREL OLIVIER. Do Atrial Differences in Endothelial Damage, Leukocyte and Platelet Activation, or Tissue Factor Activity Contribute to Chamber-Specific Thrombogenic Status in Patients with Atrial Fibrillation? J Cardiovasc Electrophysiol 2013; 25:266-70. [DOI: 10.1111/jce.12312] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 09/05/2013] [Accepted: 10/04/2013] [Indexed: 01/03/2023]
Affiliation(s)
- LAURENCE JESEL
- University of Strasbourg; EA7293 Stress vasculaire et tissulaire Strasbourg France
| | - THOMAS ARENTZ
- Universitäts-Herzzentrum Freiburg-Bad Krozingen; Klinik für Kardiologie und Angiologie II; Bad Krozingen Germany
| | - CLAUDIA HERRERA-SIKLODY
- Universitäts-Herzzentrum Freiburg-Bad Krozingen; Klinik für Kardiologie und Angiologie II; Bad Krozingen Germany
| | - DIETMAR TRENK
- Universitäts-Herzzentrum Freiburg-Bad Krozingen; Klinik für Kardiologie und Angiologie II; Bad Krozingen Germany
| | - FATIHA ZOBAIRI
- University of Strasbourg; EA7293 Stress vasculaire et tissulaire Strasbourg France
| | - MALAK ABBAS
- University of Strasbourg; EA7293 Stress vasculaire et tissulaire Strasbourg France
- Faculty of Sciences; Lebanon University; Hadath Lebanon
| | - REINHOLD WEBER
- Universitäts-Herzzentrum Freiburg-Bad Krozingen; Klinik für Kardiologie und Angiologie II; Bad Krozingen Germany
| | - JAN MINNERS
- Universitäts-Herzzentrum Freiburg-Bad Krozingen; Klinik für Kardiologie und Angiologie II; Bad Krozingen Germany
| | - FLORENCE TOTI
- University of Strasbourg; EA7293 Stress vasculaire et tissulaire Strasbourg France
| | - OLIVIER MOREL
- University of Strasbourg; EA7293 Stress vasculaire et tissulaire Strasbourg France
| |
Collapse
|
45
|
Ng KP, Edwards NC, Lip GY, Townend JN, Ferro CJ. Atrial Fibrillation in CKD: Balancing the Risks and Benefits of Anticoagulation. Am J Kidney Dis 2013; 62:615-32. [PMID: 23746378 DOI: 10.1053/j.ajkd.2013.02.381] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 02/21/2013] [Indexed: 12/20/2022]
|
46
|
Microparticles in atrial fibrillation: A link between cell activation or apoptosis, tissue remodelling and thrombogenicity. Int J Cardiol 2013; 168:660-9. [DOI: 10.1016/j.ijcard.2013.03.031] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Revised: 12/31/2012] [Accepted: 03/17/2013] [Indexed: 11/24/2022]
|
47
|
Sanborn MR, Thom SR, Bohman LE, Stein SC, Levine JM, Milovanova T, Maloney-Wilensky E, Frangos S, Kumar MA. Temporal dynamics of microparticle elevation following subarachnoid hemorrhage. J Neurosurg 2012; 117:579-86. [DOI: 10.3171/2012.6.jns111163] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object
Microparticles (MPs), small membrane fragments shed from various cell types, have been implicated in thrombosis, inflammation, and endothelial dysfunction. Their involvement in subarachnoid hemorrhage (SAH) and the development of cerebral infarction and clinical deterioration caused by delayed cerebral ischemia (DCI) remain ill defined. The authors sought to quantify the magnitude of elevations in MPs, delineate the temporal dynamics of elevation, and analyze the correlation between MPs and DCI in patients with SAH.
Methods
On the day of hemorrhage and on Days 1, 3, 5, 7, and 10 after hemorrhage, peripheral blood samples were drawn from 22 patients with SAH. Plasma samples were labeled with Annexin V and CD142, CD41a, CD235a, CD146, CD66b, or von Willebrand factor (vWF) and were quantified by flow cytometry. Clinical data, including the 3-month extended Glasgow Outcome Scale (GOS-E) scores, infarction as measured on MRI at 14 days after SAH, and vasospasm as measured by transcranial Doppler ultrasonography and angiography, were collected and compared with the MP burden.
Results
When averaged over time, all MP subtypes were elevated relative to controls. The CD235a+(erythrocyte)−, CD66b+(neutrophil)−, and vWF-associated MPs peaked on the day of hemorrhage and quickly declined. The CD142+(tissue factor [TF])–associated MPs and CD146+(endothelial cell)–associated MPs were significantly elevated throughout the study period. There was a strong negative correlation between TF-expressing and endothelial-derived MPs at Day 1 after SAH and the risk of infarction at Day 14 after SAH.
Conclusions
Microparticles of various subtypes are elevated following SAH; however, the temporal profile of this elevation varies by subtype. Those subtypes closely associated with thrombosis and endothelial dysfunction, for example, CD145+(TF)-associated MPs and CD146+(endothelial cell)–associated MPs, had the most durable response and demonstrated a significant negative correlation with radiographic infarction at 14 days after SAH. Levels of these MPs predict infarction as early as Day 1 post-SAH.
Collapse
Affiliation(s)
| | - Stephen R. Thom
- 4Department of Emergency Medicine, Institute for Environmental Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | - Joshua M. Levine
- 1Departments of Neurosurgery,
- 2Neurology, and
- 3Anesthesiology and Critical Care; and
| | - Tatyana Milovanova
- 4Department of Emergency Medicine, Institute for Environmental Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | - Monisha A. Kumar
- 1Departments of Neurosurgery,
- 2Neurology, and
- 3Anesthesiology and Critical Care; and
| |
Collapse
|
48
|
Cellular damage, platelet activation, and inflammatory response after pulmonary vein isolation: A randomized study comparing radiofrequency ablation with cryoablation. Heart Rhythm 2012; 9:189-96. [PMID: 21920484 DOI: 10.1016/j.hrthm.2011.09.017] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 09/09/2011] [Indexed: 11/24/2022]
|
49
|
Owen BAL, Xue A, Heit JA, Owen WG. Procoagulant activity, but not number, of microparticles increases with age and in individuals after a single venous thromboembolism. Thromb Res 2011; 127:39-46. [PMID: 21106230 PMCID: PMC3076027 DOI: 10.1016/j.thromres.2010.10.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 10/08/2010] [Accepted: 10/19/2010] [Indexed: 11/18/2022]
Abstract
The Calibrated Automated Thrombogram (CAT), a plate-based assay that measures thrombin generation and inhibition in plasma samples, is modified to measure the procoagulant activity of phospholipid associated with plasma microparticles (MP). The assay uses a tissue factor trigger without addition of 4 μM exogenous phospholipid (PL) used in the standard CAT. Calibrated with 4:1 posphatidylcholine- phosphatidylserine (PCPS) liposomes, the assay defines a median of 40 nM procoagulant phospholipid (PPL) equivalents in plasma containing MPs from 50 normal donors, with a range from 20 - 200 nM. Like the standard CAT, the modified assay detected no difference in plasma from 36 individuals with a history of a single episode of venous thromboembolism. However the male cases had double the PPL activity, as measured by rate of thrombin generation, of females; and there was a significant correlation among cases of increased thrombin generation with age. In contrast, there were no gender disparities or age correlations among control plasmas. The findings suggest that procoagulant activity of plasma microparticles, facilitated by a simplified, one-stage plate-based assay, offer a promising avenue of investigation of mechanisms and management of venous thromboembolic disorders.
Collapse
Affiliation(s)
- B A L Owen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | |
Collapse
|
50
|
Bal L, Ederhy S, Di Angelantonio E, Toti F, Zobairi F, Dufaitre G, Meuleman C, Mallat Z, Boccara F, Tedgui A, Freyssinet JM, Cohen A. Factors influencing the level of circulating procoagulant microparticles in acute pulmonary embolism. Arch Cardiovasc Dis 2010; 103:394-403. [PMID: 20800803 DOI: 10.1016/j.acvd.2010.06.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Revised: 06/15/2010] [Accepted: 06/17/2010] [Indexed: 12/21/2022]
Abstract
BACKGROUND Flow cytometry has shown levels of platelet-derived microparticles (PMPs) and endothelial-derived microparticles (EMPs) to be elevated in deep-vein thrombosis. Cardiovascular risk factors can also contribute to hypercoagulability due to circulating procoagulant microparticles (CPMPs). AIMS To investigate in a case-control study the respective contribution of pulmonary embolism and cardiovascular risk factors to the level of hypercoagulability due to CPMPs. METHODS CPMP, PMP and EMP levels were measured in 45 consecutive patients (age 67.9 +/- 11.6 years; 66.7% men) admitted to an intensive care unit for acute pulmonary embolism (APE), 45 healthy control subjects with no history of venous thromboembolism or vascular risk factors (Controls(noCVRFs)), and 45 patients with cardiovascular risk factors (Controls(CVRFs)). APE was diagnosed by spiral computed tomography or scintigraphy. CPMP levels were assessed using a prothrombinase assay on platelet-depleted plasma (results expressed as nmol/L equivalent). RESULTS CPMP levels were higher in APE patients than in Controls(noCVRFs) (medians 4.7 vs 3.2 nmol/L, interquartile ranges [IQRs] 2.9-11.1 vs 2.3-4.6 nmol/L; p=0.02). Similar results were reported for PMPs (medians 2.2 vs 1.9 nmol/L, IQRs 1.7-5.8 vs 1.4-2.4 nmol/L; p=0.02), whereas EMP levels were not significantly different. However, CPMP procoagulant activity was not significantly different in APE patients and Controls(CVRFs). CONCLUSIONS CPMPs and PMPs were significantly elevated in APE patients vs Controls(noCVRFs), but this correlation was not significant when APE patients were compared with Controls(CVRFs). Our observations highlight the importance of adjusting for the presence of cardiovascular risk factors in conditions in which microparticle levels are raised.
Collapse
Affiliation(s)
- Laurence Bal
- Cardiology Department, Saint-Antoine University and Medical School, Assistance Publique-Hôpitaux de Paris, université Pierre-et-Marie-Curie, 75571 Paris cedex 12, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|