Circulating microparticles: challenges and perspectives of flow cytometric assessment

1009 Clinical Focus Circulating microparticles: challenges and perspectives of flow cytometric assessment Eduard Shantsila1; Silvia Montoro-García1,...
Author: Nathan Cobb
7 downloads 2 Views 1MB Size
1009

Clinical Focus

Circulating microparticles: challenges and perspectives of flow cytometric assessment Eduard Shantsila1; Silvia Montoro-García1,2; Pilar Gallego1; Gregory Y. H. Lip1 1University

of Birmingham Centre for Cardiovascular Sciences, City Hospital, Birmingham, UK; 2Department of Cardiology, Hospital Universitario Virgen de la Arrixaca, University of Murcia, Spain

Summary Circulating blood microparticles are likely to play a significant role as messengers of biological information. Their accurate quantification and characterisation is challenging and needs to be carefully designed with preferable usage of fresh minimally-processed blood samples. Utilisation of flow cytometers specifically designed for analysis of small-size particles is likely to provide considerable methodological Correspondence to: Prof. Gregory Y. H. Lip University of Birmingham Centre for Cardiovascular Sciences City Hospital, Birmingham B18 7QH, UK Tel: +44 121 507 5080, Fax: +44 121 507 5907 E-mail: [email protected]

advantages and should be the preferable option. This viewpoint manuscript provides a critical summary of the key methodological aspects of microparticle analysis.

Keywords Microparticles, flow cytometry, microvesicles

Received: November 14, 2013 Accepted after minor revision: January 7, 2014 Prepublished online: February 20, 2014 doi:10.1160/TH13-11-0937 Thromb Haemost 2014; 111: 1009–1014

Note: The review process for this viewpoint article was fully handled by Christian Weber, Editor in Chief.

Introduction Complex biological systems are composed of myriads of cells with sophisticated mechanisms of information exchange and delivery of regulatory signals. These systems are largely represented by molecular and cellular levels of signalling, and various circulating cells (e.g. leukocytes and platelets) which would serve as ‘mobile messengers’ for biological signals (1). The gap between the molecular and cellular biological messengers is filled by different subcellular vesicles with potent regulatory properties (2, 3). Their regulatory functions are mediated by both surface receptors to allow interaction(s) with target cells as well as by the internal content of the vesicles, which can fuse with the cell content (3). These vesicles, usually called cellular microparticles (MPs) are constantly present in circulation and can thus be implicated in regulation of remote organs and tissues (4). Cellular MPs are usually defined as heterogeneous vesicles sized 0.1-1.0 μm in diameter, which express surface antigens characteristic of their parent cells (5). The size distinguishes MPs from smaller particles called exosomes, which have diameter of less 0.1 μm, and from larger particles, apoptotic bodies formed by fragments of cells undergoing apoptosis. Initially considered as ‘cell dust’ and by-product of cellular death/apoptosis circulating MPs, at least partly, produced in a regulated manner by living cells as part their biological activity (6). Various cells appear to possess mechanism(s) leading to generation of MPs enriched in specific types of membrane proteins and

sub-membrane molecules (7). This implies the presence of complex signalling pathways that both initiate and execute MP formation. However, the precise biological and clinical roles of MPs are remain poorly understood. This is largely due to existing limitations of available methods for MP quantification and characterisation. In this viewpoint, we aim to summarise the key issues related to the analysis of circulating MPs by flow cytometry, which is the principal tool of their assessment. The viewpoint includes some technical recommendations to keep in mind before designing the flow cytometry protocol for MP characterisation.

Size MPs are defined as membranous vesicles of arbitrary size from 0.1 to 1.0 μm (8). Size is one of the key factors defining these events in flow cytometric analysis, yet one of the main challenges for this technology. This approach has unique observer-independent power of analysis of huge number of cellular events. However, MP size is well below the range of reliable discrimination of conventional flow cytometers (about 5 μm). Whilst this equipment can provide some information on MPs sized 0.5-1.0 μm (as estimated using plastic beads) the smaller MPs are largely left beyond their scope of detection (9). Of note, comparisons of MPs ‘seen’ by atomic microscopy and conventional flow cytometers indicates that small-size MPs (i.e. smaller than 0.5 μm) represent majority of these structures and their exclusion from the analysis may present a major bias (10).

© Schattauer 2014

Thrombosis and Haemostasis 111.6/2014 Downloaded from www.thrombosis-online.com on 2017-01-28 | IP: 37.44.207.66 For personal or educational use only. No other uses without permission. All rights reserved.

1010

Shantsila et al. Circulating microparticles – a viewpoint

A commercially available system specifically designed for small particle analysis provides obvious advantages in this respect enabling reliable discrimination of small MPs (▶ Figure 1) (11). Forward scatter properties commonly used as an estimate of cell size is hardly appropriate for event of MP size range and side-scatter provides better MP discrimination (12). Light reflective properties of plastic ‘size’ beads may differ from those of MPs and may not provide a very accurate estimate of MP size (13). Analysis of recent papers (see Suppl. Table 1, available online at www.thrombosisonline.com) shows that older generations of flow cytometers with poor resolution of small event are commonly used for MP analysis. In summary, utilisation of flow cytometers designed to accurately discriminate submicron particles should be used in MP research.

Annexin V binding In contrast to cells, MPs may expose negatively charged phospholipids towards their surface. Living cells employ energy-dependent mechanisms of active shift of negatively charged phospholipids towards the inner side of their surface membranes, a process disrupted by cell death (e.g. apoptosis) (11). The observation that many MPs bind annexin V, which suggests exposure negatively charged phospholipids lead to speculation on apoptotic nature MPs. However, not all MPs bind annexin V and annexin V binding is highly dependent on Ca2+ concentrations (11, 14). Despite this, annexin V is a valuable marker of MPs of different origin, with the advantages of having a relatively high density of the target molecules on MPs and excellent reproducibility of measurements. In summary, annexin V-binding MPs should be analysed under strict control of (Ca2+) concentrations.

A

Sample handling The aim of any analysis of circulating MPs is to obtain information on their levels and characteristics as closely reflective of their in vivo status as possible. Achieving this is only possible with appropriate and timely handling of the blood sample, with particular attention paid to careful blood sample collection and anticoagulant use, sample centrifugation, timing of sample processing (i.e. temporal sample stability), sample freezing, thawing, and storage (15). Anticoagulants based on Ca+ chelation (sodium citrate or EDTA) are commonly used for MP analysis. As mentioned above assessment of annexin V-binding will require a careful replenishment of (Ca2+) concentrations. It is not clear at present whether residual phagocytic activity of monocytes/neutrophils and MP binding by leucocytes occurs in sodium citrate and EDTA-anticoagulated blood and how this can affect MP quantification. Importantly these processes of MP elimination could be counterbalanced by release of new MPs from cells contained in the sample and how this affects the net characteristics of the analysed events. Diverse protocols of blood centrifugation have been utilised for MP analysis (15). The centrifugation aims elimination erythrocytes and other circulating cells to achieve better clarity of the sample run through the flow cytometer with only MPs left to meet a laser beam. However, this approach opens a possibility of destruction of some MPs and fusion of others, and temporal stability of MPs and their phenotype ex vivo remains unclear (16-20). Centrifugation of platelet-rich plasma has been shown to remove 58-93% of MPs with some MP subsets being selectively lost (21-23). The impact of ultracentrifugation on MP recovery from platelet free plasma varies according to different reports (21-23). Indeed, centrifugation could also potentially lead to the artificial ‘generation’ of new microvesicles from some blood cells. Analysis

B

Figure 1: Quantification of apoptotic annexin V+ microparticles. A) Overall microparticle gating based on their size (side scatter); B) Gating of apoptotic annexin V+ microparticles. AnV, annexin V; MP, microparticles.

Thrombosis and Haemostasis 111.6/2014

© Schattauer 2014 Downloaded from www.thrombosis-online.com on 2017-01-28 | IP: 37.44.207.66

For personal or educational use only. No other uses without permission. All rights reserved.

Shantsila et al. Circulating microparticles – a viewpoint

of MPs from fresh whole blood samples is achievable and helps avoiding several procedure steps interfering with the MP count (24). Also, it is common to freeze MP-containing samples prior to their batched analysis. Freezing and prolonged storage affects MP quantity, with different types of MPs being preferentially increased or lost after the sample thawing, depending on storage conditions and/or analytical protocols used (11, 18, 20-21, 25). The duration of MP storage also matters with short-term and long-term freezing of the same samples reported to change MP levels in opposite directions (21). Nevertheless, MP quantification agreement from samples under different storage conditions tends to be relatively good and a review of the recent publications shows that MPs are most commonly analysed from frozen samples, with various storage duration and temperature (22). In summary, an ideal protocol of flow cytometric analysis of circulating MPs should be based on analysis of fresh whole blood sample stabilised with appropriate anticoagulant/preservative, which still needs to be established. Centrifugation and freezing of blood samples for MP analysis should be avoided.

MP quantification

Analysis of MP origin and surface epitopes

Different types of MPs

The small size of MPs poses another challenge for their flow cytometric assessment, a small surface area with relative small number of protein molecules for antibody binding. Indeed, a leukocyte with diameter of 10 μm has 100 times bigger surface than a 1 μm MP and 10,000 times bigger than 0.1 μm MPs. The surface of the smallest 0.1 μm is just about 0.12 μm2 with only half of it available for illumination by the flow cytometry laser. Consequently the number of specific molecules available for binding by antibodyflourochrome reagents is relatively small to produce bright signal. Amplification of the signal using a two stage biotin-streptavidin based staining has been successfully used to improve discrimination of MPs of specific origin (e.g. from platelets, monocytes, endothelial cells) (11). MPs tend to show a relatively high degree of non-specific binding. The problem is likely to stem from MP contamination by particles-remnants from necrotic and apoptotic cells, which cannot be eliminated from analysis. Unfortunately, the majority of published papers do not show any figures of representative flow cytometry based gating and acquisition strategies, thus preventing appreciation strengths and weaknesses of many individual study protocols. In summary, epitopes with the highest density on the parental cells should be used to define origin of MPs (e.g. CD42b for platelet-derived MPs, CD14 for monocyte-derived MPs, etc). Multiparametric analyses should be avoided unless absolutely essential. Use of signal-amplification approach should be considered. Nonspecific binding of antibodies should always be considered and evaluated before data acquisition. Representative figures of flow cytometrtc gating and acquisition strategies should be included in the published manuscripts.

Employment of high resolution flow cytometry (e.g. Apogee System) has revealed a uniform present of more than one cluster of small size events in blood samples (▶ Figure 1). The nature of these different clusters is not clear at present and may represent different types of MPs, aggregates of large proteins or other biological phenomena. Conventional flow cytometers may have suboptimal resolution for discrimination of different types of submicron particles. In summary, care should be taken to include into analysis clearly selected types of small blood particles.

Accurate MP quantification is essential for assessment of their pathophysiological and prognostic significance. Utilisation of dualplatform approach of MP quantification based on simultaneous counting of leucocytes (or their subsets), estimation of MP proportion to the number of these leukocytes, which absolute numbers are obtained from haematological analysis is not feasible for MPs due to profound difference in their size. Commercial count beads can be used, but they usually have a size which is substantially higher that the largest MPs, which will require appropriate adjustment of the flow cytometric plot scales to accommodate the count beads. This approach appears to be most commonly used in recently published studies. A volumetric approach aiming direct quantification of MPs in accurately measure sample volumes appears to be an optimal solution. In summary, a direct volumetric method is preferable for MP quantification. Use of ‘count beads’ is an alternative option if the volumetric method is not available. Quantification of MPs can be affected by dilution of the samples.

Quo vadis? Whilst this Viewpoint manuscript provides a rather critical overview of microparticle analysis, supporting information from our review of published data on methodological approaches for flow cytometric analysis of MPs is summarised in Suppl. Table 1 (available online at www.thrombosis-online.com) (26-118). It is clear that the published literature thus far contains considerable heterogeneity in methodology and in some cases, inadequate information (for example, on gating strategy, figures, assay variability, etc). Things would need to improve, and in this Viewpoint articles, we provide some suggestions on the way forward. Indeed, circulating blood MPs are likely to play significant role as messengers of biological information. We strongly believe that their accurate quantification and characterisation is challenging and needs to be carefully designed with preferable usage of fresh minimally-processed blood samples. Utilisation of flow cytometers specifically designed for analysis of small-size particles is likely to

© Schattauer 2014

Thrombosis and Haemostasis 111.6/2014 Downloaded from www.thrombosis-online.com on 2017-01-28 | IP: 37.44.207.66 For personal or educational use only. No other uses without permission. All rights reserved.

1011

1012

Shantsila et al. Circulating microparticles – a viewpoint

Key messages 1. An ideal protocol of flow cytometric analysis of circulating MPs should be based on analysis of fresh whole blood sample stabilised with appropriate anticoagulant/preservative, which still needs to be established. 2. Centrifugation and freezing of blood samples for MP analysis should be avoided. In summary, epitopes with the highest density on the parental cells should be used to define origin of MPs (e.g. CD42b for platelet-derived MPs, CD14 for monocyte-derived MPs, etc). 3. Multiparametric analyses should be avoided unless absolutely essential, use of signal-amplification approach should be considered and non-specific binding of antibodies should always be considered and evaluated before data acquisition.

provide considerable methodological advantages and should be the preferable option. Conflicts of interest

None declared.

This viewpoint article reflects the view of its author(s) and is not representative of the view of the Editorial Board or the Publishers.

References 1. Carlin LM, Stamatiades EG, Auffray C, et al. Nr4a1-Dependent Ly6C(low) Monocytes Monitor Endothelial Cells and Orchestrate Their Disposal. Cell 2013; 153: 362-375. 2. Shantsila E, Kamphuisen PW, Lip GY. Circulating microparticles in cardiovascular disease: implications for atherogenesis and atherothrombosis. J Thromb Haemost 2010; 8: 2358-2368. 3. Montoro-Garcia S, Shantsila E, Marin F, et al. Circulating microparticles: new insights into the biochemical basis of microparticle release and activity. Basic Res Cardiol 2011; 106: 911-923. 4. Jaiswal R, Luk F, Dalla PV, et al. Breast cancer-derived microparticles display tissue selectivity in the transfer of resistance proteins to cells. PLoS One 2013; 8: e61515. 5. Mause SF, Weber C. Microparticles: protagonists of a novel communication network for intercellular information exchange. Circ Res 2010; 107: 1047-1057. 6. Jimenez JJ, Jy W, Mauro LM, et al. Endothelial cells release phenotypically and quantitatively distinct microparticles in activation and apoptosis. Thromb Res 2003; 109: 175-180. 7. Vion AC, Ramkhelawon B, Loyer X, et al. Shear stress regulates endothelial microparticle release. Circ Res 2013; 112: 1323-1333. 8. Lacroix R, Robert S, Poncelet P, et al. Standardization of platelet-derived microparticle enumeration by flow cytometry with calibrated beads: results of the International Society on Thrombosis and Haemostasis SSC Collaborative workshop. J Thromb Haemost 2010; 8: 2571-2574. 9. Robert S, Poncelet P, Lacroix R, et al. Standardization of platelet-derived microparticle counting using calibrated beads and a Cytomics FC500 routine flow cytometer: a first step towards multicenter studies? J Thromb Haemost 2009; 7: 190-197. 10. Yuana Y, Oosterkamp TH, Bahatyrova S, et al. Atomic force microscopy: a novel approach to the detection of nanosized blood microparticles. J Thromb Haemost 2010; 8: 315-323.

11. Montoro-Garcia S, Shantsila E, Orenes-Pinero E, et al. An innovative flow cytometric approach for small-size platelet microparticles: influence of calcium. Thromb Haemost 2012; 108: 373-383. 12. Lawrie AS, Albanyan A, Cardigan RA, et al. Microparticle sizing by dynamic light scattering in fresh-frozen plasma. Vox Sang 2009; 96: 206-212. 13. Chandler WL, Yeung W, Tait JF. A new microparticle size calibration standard for use in measuring smaller microparticles using a new flow cytometer. J Thromb Haemost 2011; 9: 1216-1224. 14. Connor DE, Exner T, Ma DD, et al. The majority of circulating platelet-derived microparticles fail to bind annexin V, lack phospholipid-dependent procoagulant activity and demonstrate greater expression of glycoprotein Ib. Thromb Haemost 2010; 103: 1044-1052. 15. Yuana Y, Bertina RM, Osanto S. Pre-analytical and analytical issues in the analysis of blood microparticles. Thromb Haemost 2011; 105: 396-408. 16. Stagnara J, Garnache Ottou F, Angelot F, et al. Correlation between platelet-derived microparticle enumeration by flow cytometry and phospholipid-dependent procoagulant activity in microparticles: the centrifugation step matters! Thromb Haemost 2012; 107: 1185-1187. 17. Dignat-George F, Freyssinet JM, Key NS. Centrifugation is a crucial step impacting microparticle measurement. Platelets 2009; 20: 225-226; author reply 227-228. 18. Shah MD, Bergeron AL, Dong JF, et al. Flow cytometric measurement of microparticles: pitfalls and protocol modifications. Platelets 2008; 19: 365-372. 19. Lacroix R, Judicone C, Poncelet P, et al. Impact of pre-analytical parameters on the measurement of circulating microparticles: towards standardization of protocol. J Thromb Haemost 2012; 10: 437-446. 20. Dey-Hazra E, Hertel B, Kirsch T, et al. Detection of circulating microparticles by flow cytometry: influence of centrifugation, filtration of buffer, and freezing. Vasc Health Risk Manag 2010; 6: 1125-1133. 21. Chandler WL. Microparticle counts in platelet-rich and platelet-free plasma, effect of centrifugation and sample-processing protocols. Blood Coagul Fibrinolysis 2013; 24: 125-132. 22. van Ierssel SH, Van Craenenbroeck EM, Conraads VM, et al. Flow cytometric detection of endothelial microparticles (EMP): effects of centrifugation and storage alter with the phenotype studied. Thromb Res 2010; 125: 332-339. 23. Ayers L, Kohler M, Harrison P, et al. Measurement of circulating cell-derived microparticles by flow cytometry: sources of variability within the assay. Thromb Res 2011; 127: 370-377. 24. Christersson C, Johnell M, Siegbahn A. Evaluation of microparticles in whole blood by multicolour flow cytometry assay. Scand J Clin Lab Invest 2013; 73: 229-239. 25. Orozco AF, Lewis DE. Flow cytometric analysis of circulating microparticles in plasma. Cytometry A 2010; 77: 502-514. 26. Eleftheriou D, Ganesan V, Hong Y, et al. Endothelial injury in childhood stroke with cerebral arteriopathy: a cross-sectional study. Neurology 2012; 79: 2089-2096. 27. Larson MC, Woodliff JE, Hillery CA, et al. Phosphatidylethanolamine is externalized at the surface of microparticles. Biochim Biophys Acta 2012; 1821: 1501-1507. 28. Jeanneteau J, Hibert P, Martinez MC, et al. Microparticle release in remote ischemic conditioning mechanism. Am J Physiol Heart Circ Physiol 2012; 303: H871-877. 29. Tual-Chalot S, Fatoumata K, Priou P, et al. Circulating microparticles from patients with obstructive sleep apnea enhance vascular contraction: mandatory role of the endothelium. Am J Pathol 2012; 181: 1473-1482. 30. Petrozella L, Mahendroo M, Timmons B, et al. Endothelial microparticles and the antiangiogenic state in preeclampsia and the postpartum period. Am J Obstet Gynecol 2012; 207: 140 e20-26. 31. Singh N, Van Craeyveld E, Tjwa M, et al. Circulating apoptotic endothelial cells and apoptotic endothelial microparticles independently predict the presence of cardiac allograft vasculopathy. J Am Coll Cardiol 2012; 60: 324-331. 32. Visovatti SH, Hyman MC, Bouis D, et al. Increased CD39 nucleotidase activity on microparticles from patients with idiopathic pulmonary arterial hypertension. PLoS One 2012; 7: e40829. 33. Chen Y, Feng B, Li X, et al. Plasma endothelial microparticles and their correlation with the presence of hypertension and arterial stiffness in patients with type 2 diabetes. J Clin Hypertens 2012; 14: 455-460.

Thrombosis and Haemostasis 111.6/2014

© Schattauer 2014 Downloaded from www.thrombosis-online.com on 2017-01-28 | IP: 37.44.207.66

For personal or educational use only. No other uses without permission. All rights reserved.

Shantsila et al. Circulating microparticles – a viewpoint 34. Basavaraj MG, Olsen JO, Osterud B, et al. Differential ability of tissue factor antibody clones on detection of tissue factor in blood cells and microparticles. Thromb Res 2012; 130: 538-546. 35. Biasucci LM, Porto I, Di Vito L, et al. Differences in microparticle release in patients with acute coronary syndrome and stable angina. Circ J 2012; 76: 2174-2182. 36. Vikerfors A, Mobarrez F, Bremme K, et al. Studies of microparticles in patients with the antiphospholipid syndrome (APS). Lupus 2012; 21: 802-805. 37. Lukasik M, Michalak S, Dworacki G, et al. Reactive leptin resistance and the profile of platelet activation in acute ischaemic stroke patients. Thromb Haemost 2012; 108: 107-118. 38. Rank A, Nieuwland R, Roesner S, et al. Climacteric lowers plasma levels of platelet-derived microparticles: a pilot study in pre- versus postmenopausal women. Acta Haematol 2012; 128: 53-59. 39. Artoni A, Merati G, Padovan L, et al. Residual platelets are the main determinants of microparticles count in frozen-thawed plasma. Thromb Res 2012; 130: 561-562. 40. Lee ST, Chu K, Jung KH, et al. Circulating CD62E+ microparticles and cardiovascular outcomes. PLoS One 2012; 7: e35713. 41. Van Der Meijden PE, Van Schilfgaarde M, Van Oerle R, et al. Platelet- and erythrocyte-derived microparticles trigger thrombin generation via factor XIIa. J Thromb Haemost 2012; 10: 1355-1362. 42. Gerotziafas GT, Van Dreden P, Chaari M, et al. The acceleration of the propagation phase of thrombin generation in patients with steady-state sickle cell disease is associated with circulating erythrocyte-derived microparticles. Thromb Haemost 2012; 107: 1044-1052. 43. Katzenell S, Shomer E, Zipori Y, et al. Characterization of negatively charged phospholipids and cell origin of microparticles in women with gestational vascular complications. Thromb Res 2012; 130: 479-484. 44. Parsi K, Connor DE, Pilotelle A, et al. Low concentration detergent sclerosants induce platelet activation but inhibit aggregation due to suppression of GPIIb/ IIIa activation in vitro. Thromb Res 2012; 130: 472-478. 45. Lanuti P, Santilli F, Marchisio M, et al. A novel flow cytometric approach to distinguish circulating endothelial cells from endothelial microparticles: relevance for the evaluation of endothelial dysfunction. J Immunol Methods 2012; 380: 16-22. 46. Rautou PE, Bresson J, Sainte-Marie Y, et al. Abnormal plasma microparticles impair vasoconstrictor responses in patients with cirrhosis. Gastroenterology 2012; 143: 166-176 e6. 47. Skeppholm M, Mobarrez F, Malmqvist K, et al. Platelet-derived microparticles during and after acute coronary syndrome. Thromb Haemost 2012; 107: 1122-1129. 48. Gao C, Xie R, Yu C, et al. Procoagulant activity of erythrocytes and platelets through phosphatidylserine exposure and microparticles release in patients with nephrotic syndrome. Thromb Haemost 2012; 107: 681-689. 49. van Ierssel SH, Hoymans VY, Van Craenenbroeck EM, et al. Endothelial microparticles (EMP) for the assessment of endothelial function: an in vitro and in vivo study on possible interference of plasma lipids. PLoS One 2012; 7: e31496. 50. Thom SR, Milovanova TN, Bogush M, et al. Microparticle production, neutrophil activation, and intravascular bubbles following open-water SCUBA diving. J Appl Physiol 2012; 112: 1268-1278. 51. Park MS, Owen BA, Ballinger BA, et al. Quantification of hypercoagulable state after blunt trauma: microparticle and thrombin generation are increased relative to injury severity, while standard markers are not. Surgery 2012; 151: 831-836. 52. Walenta K, Schwarz V, Schirmer SH, et al. Circulating microparticles as indicators of peripartum cardiomyopathy. Eur Heart J 2012; 33: 1469-1479. 53. Stepien E, Stankiewicz E, Zalewski J, et al. Number of microparticles generated during acute myocardial infarction and stable angina correlates with platelet activation. Arch Med Res 2012; 43: 31-35. 54. Sari I, Bozkaya G, Kirbiyik H, et al. Evaluation of circulating endothelial and platelet microparticles in men with ankylosing spondylitis. J Rheumatol 2012; 39: 594-599. 55. Jung C, Sorensson P, Saleh N, et al. Circulating endothelial and platelet derived microparticles reflect the size of myocardium at risk in patients with ST-elevation myocardial infarction. Atherosclerosis 2012; 221: 226-231. 56. Nielsen CT, Ostergaard O, Stener L, et al. Increased IgG on cell-derived plasma microparticles in systemic lupus erythematosus is associated with autoantibodies and complement activation. Arthritis Rheum 2012; 64: 1227-1236.

57. Ge S, Hertel B, Emden SH, et al. Microparticle generation and leucocyte death in Shiga toxin-mediated HUS. Nephrol Dial Transplant 2012; 27: 2768-2775. 58. Franca CN, Pinheiro LF, Izar MC, et al. Endothelial progenitor cell mobilization and platelet microparticle release are influenced by clopidogrel plasma levels in stable coronary artery disease. Circ J 2012; 76: 729-736. 59. Holtom E, Usherwood JR, Macey MG, et al. Microparticle formation after coculture of human whole blood and umbilical artery in a novel in vitro model of flow. Cytometry A 2012; 81: 390-399. 60. Mayne E, Funderburg NT, Sieg SF, et al. Increased platelet and microparticle activation in HIV infection: upregulation of P-selectin and tissue factor expression. J Acquir Immune Defic Syndr 2012; 59: 340-346. 61. Zwicker JI, Lacroix R, Dignat-George F, et al. Measurement of platelet microparticles. Methods Mol Biol 2012; 788: 127-139. 62. Ye R, Ye C, Huang Y, et al. Circulating tissue factor positive microparticles in patients with acute recurrent deep venous thrombosis. Thromb Res 2012; 130: 253-258. 63. Jayachandran M, Miller VM, Heit JA, et al. Methodology for isolation, identification and characterization of microvesicles in peripheral blood. J Immunol Methods 2012; 375: 207-214. 64. Emmerechts J, Jacobs L, Van Kerckhoven S, et al. Air pollution-associated procoagulant changes: the role of circulating microvesicles. J Thromb Haemost 2012; 10: 96-106. 65. Amabile N, Guerin AP, Tedgui A, et al. Predictive value of circulating endothelial microparticles for cardiovascular mortality in end-stage renal failure: a pilot study. Nephrol Dial Transplant 2012; 27: 1873-1880. 66. Mobarrez F, Egberg N, Antovic J, et al. Release of endothelial microparticles in vivo during atorvastatin treatment; a randomized double-blind placebo-controlled study. Thromb Res 2012; 129: 95-97. 67. Fink K, Feldbrugge L, Schwarz M, et al. Circulating annexin V positive microparticles in patients after successful cardiopulmonary resuscitation. Crit Care 2011; 15: R251. 68. Alijotas-Reig J, Palacio-Garcia C, Farran-Codina I, et al. Circulating cell-derived microparticles in severe preeclampsia and in fetal growth restriction. Am J Reprod Immunol 2012; 67: 140-151. 69. Aass HC, Ovstebo R, Troseid AM, et al. Fluorescent particles in the antibody solution result in false TF- and CD14-positive microparticles in flow cytometric analysis. Cytometry A 2011; 79: 990-999. 70. Martinez-Sales V, Vila V, Mico L, et al. Circulating endothelial cells and microparticles in patients with antiphospholipid antibodies. Med Clin 2011; 136: 431-433. 71. Burdess A, Michelsen AE, Brosstad F, et al. Platelet activation in patients with peripheral vascular disease: reproducibility and comparability of platelet markers. Thromb Res 2012; 129: 50-55. 72. Wu Q, Chen H, Fang J, et al. Elevated Fas/FasL system and endothelial cell microparticles are involved in endothelial damage in acute graft-versus-host disease: a clinical analysis. Leuk Res 2012; 36: 275-280. 73. Csongradi E, Nagy B, Jr., Fulop T, et al. Increased levels of platelet activation markers are positively associated with carotid wall thickness and other atherosclerotic risk factors in obese patients. Thromb Haemost 2011; 106: 683-692. 74. Perez-Casal M, Thompson V, Downey C, et al. The clinical and functional relevance of microparticles induced by activated protein C treatment in sepsis. Crit Care 2011; 15: R195. 75. Hayashi M, Takeshita K, Inden Y, et al. Platelet activation and induction of tissue factor in acute and chronic atrial fibrillation: involvement of mononuclear cell-platelet interaction. Thromb Res 2011; 128: e113-118. 76. Lincz LF, Scorgie FE, Enjeti A, et al. Variable plasma levels of Factor V Leiden correlate with circulating platelet microparticles in carriers of Factor V Leiden. Thromb Res 2012; 129: 192-196. 77. La Vignera S, Condorelli R, D'Agata R, et al. Dysfunction of the endothelial-platelet pathway in patients with erectile dysfunction before and after daily treatment with tadalafil. Andrologia 2012; 44: 152-156. 78. Zahra S, Anderson JA, Stirling D, et al. Plasma microparticles are not elevated in fresh plasma from patients with gynaecological malignancy--an observational study. Gynecol Oncol 2011; 123: 152-156. 79. Lee RD, Barcel DA, Williams JC, et al. Pre-analytical and analytical variables affecting the measurement of plasma-derived microparticle tissue factor activity. Thromb Res 2012; 129: 80-85.

© Schattauer 2014

Thrombosis and Haemostasis 111.6/2014 Downloaded from www.thrombosis-online.com on 2017-01-28 | IP: 37.44.207.66 For personal or educational use only. No other uses without permission. All rights reserved.

1013

1014

Shantsila et al. Circulating microparticles – a viewpoint 80. Macey M, Hagi-Pavli E, Stewart J, et al. Age, gender and disease-related platelet and neutrophil activation ex vivo in whole blood samples from patients with Behcet's disease. Rheumatology 2011; 50: 1849-1859. 81. Faille D, Frere C, Cuisset T, et al. CD11b+ leukocyte microparticles are associated with high-risk angiographic lesions and recurrent cardiovascular events in acute coronary syndromes. J Thromb Haemost 2011; 9: 1870-1873. 82. Nielsen CT, Ostergaard O, Johnsen C, et al. Distinct features of circulating microparticles and their relationship to clinical manifestations in systemic lupus erythematosus. Arthritis Rheum 2011; 63: 3067-3077. 83. De Rop C, Stadler M, Buchholz S, et al. Evaluation of tissue factor bearing microparticles as biomarkers in allogeneic stem-cell transplantation. Transplantation 2011; 92: 351-358. 84. da Silva EF, Fonseca FA, Franca CN, et al. Imbalance between endothelial progenitors cells and microparticles in HIV-infected patients naive for antiretroviral therapy. AIDS 2011; 25: 1595-1601. 85. Tamburrelli C, Crescente M, Izzi B, et al. Epoprostenol inhibits human plateletleukocyte mixed conjugate and platelet microparticle formation in whole blood. Thromb Res 2011; 128: 446-451. 86. Mobarrez F, He S, Broijersen A, et al. Atorvastatin reduces thrombin generation and expression of tissue factor, P-selectin and GPIIIa on platelet-derived microparticles in patients with peripheral arterial occlusive disease. Thromb Haemost 2011; 106: 344-352. 87. Ramacciotti E, Blackburn S, Hawley AE, et al. Evaluation of soluble P-selectin as a marker for the diagnosis of deep venous thrombosis. Clin Appl Thromb Hemost 2011; 17: 425-431. 88. Lowery-Nordberg M, Eaton E, Gonzalez-Toledo E, et al. The effects of high dose interferon-beta1a on plasma microparticles: correlation with MRI parameters. J Neuroinflammation 2011; 8: 43. 89. Koiou E, Tziomalos K, Katsikis I, et al. Circulating platelet-derived microparticles are elevated in women with polycystic ovary syndrome diagnosed with the 1990 criteria and correlate with serum testosterone levels. Eur J Endocrinol 2011; 165: 63-68. 90. Trzepizur W, Priou P, Paris A, et al. Nocturnal release of leukocyte-derived microparticles in males with obstructive sleep apnoea. Eur Respir J 2011; 37: 1293-1295. 91. Martel C, Cointe S, Maurice P, et al. Requirements for membrane attack complex formation and anaphylatoxins binding to collagen-activated platelets. PLoS One 2011; 6: e18812. 92. Sadallah S, Eken C, Martin PJ, et al. Microparticles (ectosomes) shed by stored human platelets downregulate macrophages and modify the development of dendritic cells. J Immunol 2011; 186: 6543-6552. 93. Gordon C, Gudi K, Krause A, et al. Circulating endothelial microparticles as a measure of early lung destruction in cigarette smokers. Am J Respir Crit Care Med 2011; 184: 224-232. 94. Matijevic N, Wang YW, Kostousov V, et al. Decline in platelet microparticles contributes to reduced hemostatic potential of stored plasma. Thromb Res 2011; 128: 35-41. 95. Pelletier F, Garnache-Ottou F, Angelot F, et al. Increased levels of circulating endothelial-derived microparticles and small-size platelet-derived microparticles in psoriasis. J Invest Dermatol 2011; 131: 1573-1576. 96. Schweintzger S, Schlagenhauf A, Leschnik B, et al. Microparticles in newborn cord blood: slight elevation after normal delivery. Thromb Res 2011; 128: 62-67. 97. Rank A, Nieuwland R, Delker R, et al. Surveillance of megakaryocytic function by measurement of CD61-exposing microparticles in allogeneic hematopoietic stem cell recipients. Clin Transplant 2011; 25: E233-242. 98. Bulut D, Scheeler M, Niedballa LM, et al. Effects of immunoadsorption on endothelial function, circulating endothelial progenitor cells and circulating microparticles in patients with inflammatory dilated cardiomyopathy. Clin Res Cardiol 2011; 100: 603-610.

99. Hamilos M, Muller O, Ntalianis A, et al. Relationship between peripheral arterial reactive hyperemia and residual platelet reactivity after 600 mg clopidogrel. J Thromb Thrombolysis 2011; 32: 64-71. 100. Alkhatatbeh MJ, Mhaidat NM, Enjeti AK, et al. The putative diabetic plasma marker, soluble CD36, is non-cleaved, non-soluble and entirely associated with microparticles. J Thromb Haemost 2011; 9: 844-851. 101. Alijotas-Reig J, Palacio-Garcia C, Farran-Codina I, et al. Circulating cell-derived microparticles in women with pregnancy loss. Am J Reprod Immunol 2011; 66: 199-208. 102. Kim J, Bhattacharjee R, Kheirandish-Gozal L, et al. Circulating microparticles in children with sleep disordered breathing. Chest 2011; 140: 408-417. 103. Trappenburg MC, van Schilfgaarde M, Bredewold EO, et al. Elevated numbers and altered subsets of procoagulant microparticles in breast cancer patients using endocrine therapy. Thromb Res 2011; 127: 363-369. 104. Campello E, Spiezia L, Radu CM, et al. Endothelial, platelet, and tissue factorbearing microparticles in cancer patients with and without venous thromboembolism. Thromb Res 2011; 127: 473-477. 105. Reyna-Villasmil E, Mejia-Montilla J, Reyna-Villasmil N, et al. Endothelial microparticles in preeclampsia and eclampsia. Med Clin 2011; 136: 522-526. 106. Rank A, Nieuwland R, Crispin A, et al. Clearance of platelet microparticles in vivo. Platelets 2011; 22: 111-116. 107. Sinning JM, Losch J, Walenta K, et al. Circulating CD31+/Annexin V+ microparticles correlate with cardiovascular outcomes. Eur Heart J 2011; 32: 2034-2041. 108. Sossdorf M, Otto GP, Claus RA, et al. Cell-derived microparticles promote coagulation after moderate exercise. Med Sci Sports Exerc 2011; 43: 1169-1176. 109. Keating FK, Butenas S, Fung MK, et al. Platelet-white blood cell (WBC) interaction, WBC apoptosis, and procoagulant activity in stored red blood cells. Transfusion 2011; 51: 1086-1095. 110. Marin C, Ramirez R, Delgado-Lista J, et al. Mediterranean diet reduces endothelial damage and improves the regenerative capacity of endothelium. Am J Clin Nutr 2011; 93: 267-274. 111. Hou J, Fu Y, Zhou J, et al. Lactadherin functions as a probe for phosphatidylserine exposure and as an anticoagulant in the study of stored platelets. Vox Sang 2011; 100: 187-195. 112. Xu Y, Nakane N, Maurer-Spurej E. Novel test for microparticles in platelet-rich plasma and platelet concentrates using dynamic light scattering. Transfusion 2011; 51: 363-370. 113. Rank A, Nieuwland R, Liebhardt S, et al. Apheresis platelet concentrates contain platelet-derived and endothelial cell-derived microparticles. Vox Sang 2011; 100: 179-186. 114. Diehl P, Aleker M, Helbing T, et al. Increased platelet, leukocyte and endothelial microparticles predict enhanced coagulation and vascular inflammation in pulmonary hypertension. J Thromb Thrombolysis 2011; 31: 173-179. 115. Macey MG, Enniks N, Bevan S. Flow cytometric analysis of microparticle phenotype and their role in thrombin generation. Cytometry B Clin Cytom 2011; 80: 57-63. 116. Dursun I, Dusunsel R, Poyrazoglu HM, et al. Circulating endothelial microparticles in children with Henoch-Schonlein purpura; preliminary results. Rheumatol Int 2011; 31: 1595-1600. 117. Helal O, Defoort C, Robert S, et al. Increased levels of microparticles originating from endothelial cells, platelets and erythrocytes in subjects with metabolic syndrome: relationship with oxidative stress. Nutr Metab Cardiovasc Dis 2011; 21: 665-671. 118. Montoro-Garcia S, Shantsila E, Tapp LD, et al. Small-size circulating microparticles in acute coronary syndromes: relevance to fibrinolytic status, reparative markers and outcomes. Atherosclerosis 2013; 227: 313-322.

Thrombosis and Haemostasis 111.6/2014

© Schattauer 2014 Downloaded from www.thrombosis-online.com on 2017-01-28 | IP: 37.44.207.66

For personal or educational use only. No other uses without permission. All rights reserved.

Suggest Documents