101
|
Roberts-Dalton HD, Cocks A, Falcon-Perez JM, Sayers EJ, Webber JP, Watson P, Clayton A, Jones AT. Fluorescence labelling of extracellular vesicles using a novel thiol-based strategy for quantitative analysis of cellular delivery and intracellular traffic. NANOSCALE 2017; 9:13693-13706. [PMID: 28880029 DOI: 10.1039/c7nr04128d] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Extracellular vesicles, including exosomes, are naturally derived nanovesicles generated in and released by numerous cell types. As extracellular entities they have the capacity to interact with neighbouring cells and distant tissues and affect physiological processes as well as being implicated in numerous diseases including tumorigenesis and neurodegeneration. They are also under intense investigation as delivery vectors for biotherapeutics. The ways in which EVs interact with recipient cells to influence cell physiology and deliver a macromolecular payload are at the early stages of exploration. A significant challenge within these studies is the ability to label EVs directly or indirectly with fluorescent probes to allow visualization without compromising functionality. Here, we present a thiol-based fluorescence labelling method allowing comprehensive analysis of the cellular uptake of prostate cancer derived EVs in live cells using confocal microscopy. Labelling of the EVs in this way did not influence their size and had no effect on their ability to induce differentiation of lung fibroblasts to myofibroblasts. For endocytosis analyses, depletion of key endocytic proteins and the use of chemical inhibitors (Dynasore, EIPA, Rottlerin and IPA-3) indicated that fluid-phase endocytosis and/or macropinocytosis was involved in EV internalisation. Over a period of six hours EVs were observed to increasingly co-localise with lysosomes, indicating a possible termination point following internalisation. Overall this method provides new opportunities for analysing the cellular dynamics of EVs as biological entities affecting cell and whole body physiology as well as investigating their potential as drug delivery vectors.
Collapse
Affiliation(s)
- H D Roberts-Dalton
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, CF10 3NB, UK.
| | | | | | | | | | | | | | | |
Collapse
|
102
|
Thrombin-derived host defence peptide modulates neutrophil rolling and migration in vitro and functional response in vivo. Sci Rep 2017; 7:11201. [PMID: 28894159 PMCID: PMC5593972 DOI: 10.1038/s41598-017-11464-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/24/2017] [Indexed: 01/24/2023] Open
Abstract
Host defence peptides (HDPs) derived from the C-terminus of thrombin are proteolytically generated by enzymes released during inflammation and wounding. In this work, we studied the effects of the prototypic peptide GKY25 (GKYGFYTHVFRLKKWIQKVIDQFGE), on neutrophil functions. In vitro, GKY25 was shown to decrease LPS-induced neutrophil activation. In addition, the peptide induced CD62L shedding on neutrophils without inducing their activation. Correspondingly, GKY25-treated neutrophils showed reduced attachment and rolling behaviour on surfaces coated with the CD62L ligand E-selectin. The GKY25-treated neutrophils also displayed a dampened chemotactic response against the chemokine IL-8. Furthermore, in vivo, mice treated with GKY25 exhibited a reduced local ROS response against LPS. Taken together, our results show that GKY25 can modulate neutrophil functions in vitro and in vivo.
Collapse
|
103
|
A hybrid resistive pulse-optical detection platform for microfluidic experiments. Sci Rep 2017; 7:10173. [PMID: 28860641 PMCID: PMC5579027 DOI: 10.1038/s41598-017-10000-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/01/2017] [Indexed: 12/31/2022] Open
Abstract
Resistive-pulse sensing is a label-free method for characterizing individual particles as they pass through ion-conducting channels or pores. During a resistive pulse experiment, the ionic current through a conducting channel is monitored as particles suspended in the solution translocate through the channel. The amplitude of the current decrease during a translocation, or ‘pulse’, depends not only on the ratio of the particle and channel sizes, but also on the particle position, which is difficult to resolve with the resistive pulse signal alone. We present experiments of simultaneous electrical and optical detection of particles passing through microfluidic channels to resolve the positional dependencies of the resistive pulses. Particles were tracked simultaneously in the two signals to create a mapping of the particle position to resistive pulse amplitude at the same instant in time. The hybrid approach will improve the accuracy of object characterization and will pave the way for observing dynamic changes of the objects such as deformation or change in orientation. This combined approach of optical detection and resistive pulse sensing will join with other attempts at hybridizing high-throughput detection techniques such as imaging flow cytometry.
Collapse
|
104
|
Abstract
BACKGROUND Allogeneic mesenchymal stem cells (MSCs) show great potential for the treatment of military and civilian trauma based on their reduced immunogenicity and ability to modulate inflammation and immune function in the recipient. Although generally considered to be safe, MSCs express tissue factor (TF), a potent activator of coagulation. In the current study, we evaluated multiple MSC populations for tissue factor expression and procoagulant activity to characterize safety considerations for systemic use of MSCs in trauma patients who may have altered coagulation homeostasis. METHODS Multiple MSC populations derived from either human adipose tissue or bone marrow were expanded in the recommended stem cell media. Stem cell identity was confirmed using a well-characterized panel of positive and negative markers. Tissue factor expression on the cell surface was evaluated by flow cytometry with anti-CD142 antibody. Effects on blood coagulation were determined by thromboelastography and calibrated automated thrombogram assays using platelet-poor plasma or whole blood. RESULTS Mesenchymal stem cells express tissue factor on their surfaces and are procoagulant in the presence of blood or plasma. The adipose-derived MSCs (Ad-MSC) evaluated were more procoagulant and expressed more tissue factor than bone marrow MSCs (BM-MSCs), which showed a greater variability in TF expression. Bone marrow MSCs were identified that exhibited low procoagulant activity, whereas all Ad-MSCs examined exhibited high procoagulant activity. The percentage of cells in a given population expressing surface tissue factor correlates roughly with functional procoagulant activity. Mesenchymal stem cell tissue factor expression and procoagulant activity change over time in culture. CONCLUSIONS All MSC populations are not equivalent; care should be taken to select cells for clinical use that minimize potential safety problems and maximize chance of patient benefit. Adipose-derived MSCs seem more consistently procoagulant than BM-MSCs, presenting a potential safety concern for systemic administration in coagulopathic patients. Donor variation exists between different cell populations, and culture handling conditions may also determine coagulation activity. Cells must be routinely monitored during preparation to ensure that they retain the desired characteristics before patient administration.
Collapse
|
105
|
The ESCRT-III pathway facilitates cardiomyocyte release of cBIN1-containing microparticles. PLoS Biol 2017; 15:e2002354. [PMID: 28806752 PMCID: PMC5570487 DOI: 10.1371/journal.pbio.2002354] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 08/24/2017] [Accepted: 07/26/2017] [Indexed: 12/17/2022] Open
Abstract
Microparticles (MPs) are cell–cell communication vesicles derived from the cell surface plasma membrane, although they are not known to originate from cardiac ventricular muscle. In ventricular cardiomyocytes, the membrane deformation protein cardiac bridging integrator 1 (cBIN1 or BIN1+13+17) creates transverse-tubule (t-tubule) membrane microfolds, which facilitate ion channel trafficking and modulate local ionic concentrations. The microfold-generated microdomains continuously reorganize, adapting in response to stress to modulate the calcium signaling apparatus. We explored the possibility that cBIN1-microfolds are externally released from cardiomyocytes. Using electron microscopy imaging with immunogold labeling, we found in mouse plasma that cBIN1 exists in membrane vesicles about 200 nm in size, which is consistent with the size of MPs. In mice with cardiac-specific heterozygous Bin1 deletion, flow cytometry identified 47% less cBIN1-MPs in plasma, supporting cardiac origin. Cardiac release was also evidenced by the detection of cBIN1-MPs in medium bathing a pure population of isolated adult mouse cardiomyocytes. In human plasma, osmotic shock increased cBIN1 detection by enzyme-linked immunosorbent assay (ELISA), and cBIN1 level decreased in humans with heart failure, a condition with reduced cardiac muscle cBIN1, both of which support cBIN1 release in MPs from human hearts. Exploring putative mechanisms of MP release, we found that the membrane fission complex endosomal sorting complexes required for transport (ESCRT)-III subunit charged multivesicular body protein 4B (CHMP4B) colocalizes and coimmunoprecipitates with cBIN1, an interaction enhanced by actin stabilization. In HeLa cells with cBIN1 overexpression, knockdown of CHMP4B reduced the release of cBIN1-MPs. Using truncation mutants, we identified that the N-terminal BAR (N-BAR) domain in cBIN1 is required for CHMP4B binding and MP release. This study links the BAR protein superfamily to the ESCRT pathway for MP biogenesis in mammalian cardiac ventricular cells, identifying elements of a pathway by which cytoplasmic cBIN1 is released into blood. Microparticles are small vesicles generated from the cell surface membrane and externally released for communication with other cells. We now show that heart ventricular muscle cells, which form the main pumping chambers of the heart, release microparticles in both mouse and human. Ventricular microparticles arise from surface membrane microdomains organized by cardiac bridging integrator 1 (cBIN1), a membrane deformation protein that has been shown to be reduced during human heart failure. Here we identify microparticles containing cBIN1 in blood, which were reduced in mutant mice lacking heart cBIN1 expression. Furthermore, the process leading to microparticle release involves the recruitment of CHMP4B protein to snip the cBIN1 membrane. In humans, cBIN1 is present in blood and within microparticles. Upon osmotic shock, human microparticles burst, allowing for the quantification of cBIN1 in plasma by enzyme-linked immunosorbent assay (ELISA). The measured cBIN1 level was greatly reduced in patients with heart failure. Thus, we introduce the biology for a new blood-based diagnostic tool that can assess cardiac muscle health and identify failing heart in human patients.
Collapse
|
106
|
Probst C, Zeng Y, Zhu RR. Characterization of Protein Particles in Therapeutic Formulations Using Imaging Flow Cytometry. J Pharm Sci 2017; 106:1952-1960. [DOI: 10.1016/j.xphs.2017.04.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/12/2017] [Accepted: 04/17/2017] [Indexed: 01/18/2023]
|
107
|
Greven J, Pfeifer R, Zhi Q, Pape HC. Update on the role of endothelial cells in trauma. Eur J Trauma Emerg Surg 2017; 44:667-677. [PMID: 28674817 DOI: 10.1007/s00068-017-0812-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 06/21/2017] [Indexed: 12/23/2022]
Abstract
PURPOSE This review gives an overview of physiological processes, mainly regarding vascular endothelial cells and their important role in hemostasis, information processing, and communication during trauma. An insight is given into molecules and cells involved in the first innate immune response through to the behavior of endothelial cells in developing trauma. The goal of this review is to show the overlap of crucial factors related to the endothelium and the development of trauma. METHODS A systemic literature search was performed using Google scholar and PubMed. RESULTS The results of the literature search showed that the endothelium, especially the vascular endothelium, is involved in various cellular and subcellular pathways of activation, suppression, and transfer of information. A variety of molecules and cells are orchestrated, subsequently the endothelium gets in contact with a traumatizing event. CONCLUSION The endothelium is one of the first barriers that comes into contact with exo- and endogenous trauma-related signals and is a pivotal point in activating subsequent pathways and cascades by transfer of information.
Collapse
Affiliation(s)
- J Greven
- Department of Trauma and Reconstructive Surgery, University of Aachen Medical Center, Pauwelsstr 30, 52074, Aachen, Germany.
| | - R Pfeifer
- Department for Traumatology, University of Zürich Medical Center, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Q Zhi
- Department of Trauma and Reconstructive Surgery, University of Aachen Medical Center, Pauwelsstr 30, 52074, Aachen, Germany
| | - H C Pape
- Department for Traumatology, University of Zürich Medical Center, Rämistrasse 100, 8091, Zurich, Switzerland
| |
Collapse
|
108
|
Szabó-Taylor KÉ, Tóth EÁ, Balogh AM, Sódar BW, Kádár L, Pálóczi K, Fekete N, Németh A, Osteikoetxea X, Vukman KV, Holub M, Pállinger É, Nagy G, Winyard PG, Buzás EI. Monocyte activation drives preservation of membrane thiols by promoting release of oxidised membrane moieties via extracellular vesicles. Free Radic Biol Med 2017; 108:56-65. [PMID: 28323130 DOI: 10.1016/j.freeradbiomed.2017.03.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/06/2017] [Accepted: 03/14/2017] [Indexed: 12/15/2022]
Abstract
The redox state of cellular exofacial molecules is reflected by the amount of available thiols. Furthermore, surface thiols can be considered as indicators of immune cell activation. One group of thiol containing proteins, peroxiredoxins, in particular, have been associated with inflammation. In this study, we assessed surface thiols of the U937 and Thp1 monocyte cell lines and primary monocytes in vitro upon inflammatory stimulation by irreversibly labelling the cells with a fluorescent derivative of maleimide. We also investigated exofacial thiols on circulating blood mononuclear cells in patients with rheumatoid arthritis and healthy controls. When analysing extracellular vesicles, we combined thiol labelling with the use of antibodies to specific CD markers to exclude extracellular vesicle mimicking signals from thiol containing protein aggregates. Furthermore, differential detergent lysis was applied to confirm the vesicular nature of the detected extracellular events in blood plasma. We found an increase in exofacial thiols on monocytes upon in vitro stimulation by LPS or TNF, both in primary monocytes and monocytic cell lines (p<0.0005). At the same time, newly released extracellular vesicles showed a decrease in their exofacial thiols compared with those from unstimulated cells (p<0.05). We also found a significant elevation of surface thiols on circulating monocytes in rheumatoid arthritis patients (p<0.05) and newly released extracellular vesicles of isolated CD14+ cells from rheumatoid arthritis patients had decreased thiol levels compared with healthy subjects (p<0.01). Exofacial peroxiredoxin 1 was demonstrated on the surface of primary and cultured monocytes, and the number of peroxiredoxin 1 positive extracellular vesicles was increased in rheumatoid arthritis blood plasma (p<0.05). Furthermore, an overoxidised form of peroxiredoxin was detected in extracellular vesicle-enriched preparations from blood plasma. Our data show that cell surface thiols play a protective role and reflect oxidative stress resistance state in activated immune cells. Furthermore, they support a role of extracellular vesicles in the redox regulation of human monocytes, possibly representing an antioxidant mechanism.
Collapse
Affiliation(s)
- K É Szabó-Taylor
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary.
| | - E Á Tóth
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - A M Balogh
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - B W Sódar
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - L Kádár
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - K Pálóczi
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - N Fekete
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - A Németh
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - X Osteikoetxea
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - K V Vukman
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - M Holub
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - É Pállinger
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - Gy Nagy
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary; Semmelweis University, Department of Rheumatology, 3rd Department of Internal Medicine, 1023 Budapest, Hungary
| | - P G Winyard
- University of Exeter Medical School, St Luke's Campus, Heavitree Road, Exeter EX1 2LU, United Kingdom
| | - E I Buzás
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| |
Collapse
|
109
|
M2 Monocyte Microparticles Are Increased in Intracerebral Hemorrhage. J Stroke Cerebrovasc Dis 2017; 26:2369-2375. [PMID: 28606659 DOI: 10.1016/j.jstrokecerebrovasdis.2017.05.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 05/17/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a severe neurologic condition with no proven treatment. Recent evidence suggests that monocytes, a heterogenous group of cells with M1 and M2 phenotypes, contribute to secondary damage following ICH. Microparticles are vesicles .1-1 µm in size that are released from cells. We hypothesized that M1 and M2 monocyte microparticles (mMP) would be differentially expressed in ICH cases and controls. METHODS In a single-center, prospective, observational study, consecutive ICH cases were enrolled within 12 hours of symptom onset. Age (±5 years)-, race-, and sex-matched controls were recruited. M1 and M2 mMP numbers were determined in plasma samples using flow cytometry and protein biomarkers using standardized assays. The Mann-Whitney U test compared M1 and M2 mMP counts between cases and controls. Standardized regression coefficients compared M1 and M2 mMP with C-reactive protein (CRP) and serum amyloid A (SAA). RESULTS Nineteen ICH case-control pairs were enrolled. The median number of M1 mMP was not significantly different between ICH cases (8.63 × 107/milliliter (mL)) compared with controls (8.64 × 107/mL), (P = .525). The median number of M2 mMP was significantly higher in ICH cases (1.61 × 106/mL) compared with controls (4.46 × 105/mL) (P = .027). There were no significant associations for M1 or M2 mMP with CRP or SAA. CONCLUSION Higher numbers of M2 mMP in ICH cases compared with controls is hypothesis generating. It may represent differences in the chronic inflammatory status in patients susceptible to ICH, such as cellular activation or apoptosis. Further research is needed, including serial plasma samples, to elucidate the pathophysiology of monocytes and mMP following ICH.
Collapse
|
110
|
The Methods of Choice for Extracellular Vesicles (EVs) Characterization. Int J Mol Sci 2017; 18:ijms18061153. [PMID: 28555055 PMCID: PMC5485977 DOI: 10.3390/ijms18061153] [Citation(s) in RCA: 365] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 11/24/2022] Open
Abstract
In recent years, extracellular vesicles (EVs) have become a subject of intense study. These membrane-enclosed spherical structures are secreted by almost every cell type and are engaged in the transport of cellular content (cargo) from parental to target cells. The impact of EVs transfer has been observed in many vital cellular processes including cell-to-cell communication and immune response modulation; thus, a fast and precise characterization of EVs may be relevant for both scientific and diagnostic purposes. In this review, the most popular analytical techniques used in EVs studies are presented with the emphasis on exosomes and microvesicles characterization.
Collapse
|
111
|
Hromada C, Mühleder S, Grillari J, Redl H, Holnthoner W. Endothelial Extracellular Vesicles-Promises and Challenges. Front Physiol 2017; 8:275. [PMID: 28529488 PMCID: PMC5418228 DOI: 10.3389/fphys.2017.00275] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/18/2017] [Indexed: 12/22/2022] Open
Abstract
Extracellular vesicles, including exosomes, microparticles, and apoptotic bodies, are phospholipid bilayer-enclosed vesicles that have once been considered as cell debris lacking biological functions. However, they have recently gained immense interest in the scientific community due to their role in intercellular communication, immunity, tissue regeneration as well as in the onset, and progression of various pathologic conditions. Extracellular vesicles of endothelial origin have been found to play a versatile role in the human body, since they are on the one hand known to contribute to cardiovascular diseases, but on the other hand have also been reported to promote endothelial cell survival. Hence, endothelial extracellular vesicles hold promising therapeutic potential to be used as a new tool to detect as well as treat a great number of diseases. This calls for clinically approved, standardized, and efficient isolation and characterization protocols to harvest and purify endothelial extracellular vesicles. However, such methods and techniques to fulfill stringent requirements for clinical trials have yet to be developed or are not harmonized internationally. In this review, recent advances and challenges in the field of endothelial extracellular vesicle research are discussed and current problems and limitations regarding isolation and characterization are pointed out.
Collapse
Affiliation(s)
- Carina Hromada
- AUVA Research Centre, Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyVienna, Austria.,Austrian Cluster for Tissue RegenerationVienna, Austria
| | - Severin Mühleder
- AUVA Research Centre, Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyVienna, Austria.,Austrian Cluster for Tissue RegenerationVienna, Austria
| | - Johannes Grillari
- Austrian Cluster for Tissue RegenerationVienna, Austria.,Christian Doppler Laboratory on Biotechnology of Skin Aging, Department of Biotechnology, University of Natural Resources and Life SciencesVienna, Austria.,Evercyte GmbHVienna, Austria
| | - Heinz Redl
- AUVA Research Centre, Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyVienna, Austria.,Austrian Cluster for Tissue RegenerationVienna, Austria
| | - Wolfgang Holnthoner
- AUVA Research Centre, Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyVienna, Austria.,Austrian Cluster for Tissue RegenerationVienna, Austria
| |
Collapse
|
112
|
Piffoux M, Silva AKA, Lugagne JB, Hersen P, Wilhelm C, Gazeau F. Extracellular Vesicle Production Loaded with Nanoparticles and Drugs in a Trade-off between Loading, Yield and Purity: Towards a Personalized Drug Delivery System. ACTA ACUST UNITED AC 2017; 1:e1700044. [DOI: 10.1002/adbi.201700044] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Max Piffoux
- Laboratoire Matière et Systèmes Complexes; UMR 7057; CNRS and Université Paris Diderot; 10 rue Alice Domon et Léonie Duquet 75205 Paris Cedex 13 France
| | - Amanda K. A. Silva
- Laboratoire Matière et Systèmes Complexes; UMR 7057; CNRS and Université Paris Diderot; 10 rue Alice Domon et Léonie Duquet 75205 Paris Cedex 13 France
| | - Jean-Baptiste Lugagne
- Laboratoire Matière et Systèmes Complexes; UMR 7057; CNRS and Université Paris Diderot; 10 rue Alice Domon et Léonie Duquet 75205 Paris Cedex 13 France
| | - Pascal Hersen
- Laboratoire Matière et Systèmes Complexes; UMR 7057; CNRS and Université Paris Diderot; 10 rue Alice Domon et Léonie Duquet 75205 Paris Cedex 13 France
| | - Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes; UMR 7057; CNRS and Université Paris Diderot; 10 rue Alice Domon et Léonie Duquet 75205 Paris Cedex 13 France
| | - Florence Gazeau
- Laboratoire Matière et Systèmes Complexes; UMR 7057; CNRS and Université Paris Diderot; 10 rue Alice Domon et Léonie Duquet 75205 Paris Cedex 13 France
| |
Collapse
|
113
|
Almizraq RJ, Seghatchian J, Holovati JL, Acker JP. Extracellular vesicle characteristics in stored red blood cell concentrates are influenced by the method of detection. Transfus Apher Sci 2017; 56:254-260. [DOI: 10.1016/j.transci.2017.03.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
114
|
Johnson SM, Dempsey C, Parker C, Mironov A, Bradley H, Saha V. Acute lymphoblastic leukaemia cells produce large extracellular vesicles containing organelles and an active cytoskeleton. J Extracell Vesicles 2017; 6:1294339. [PMID: 28386390 PMCID: PMC5373679 DOI: 10.1080/20013078.2017.1294339] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Accepted: 01/26/2016] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles have been described in non-paracrine cellular interactions in cancer. We report a similar phenomenon in B-cell precursor (BCP) acute lymphoblastic leukaemia (ALL). Using advanced microscopy and high throughput screening, we further characterise a subset of large vesicles (LEVs) identified in cell lines, murine models of human BCP-ALL and clinical samples. Primary ALL blasts and cell lines released heterogeneous anucleate vesicles <6 micron into extracellular fluids. Larger LEVs were enclosed in continuous membranes, contained intact organelles and demonstrated an organised cytoskeleton. An excess of circulating CD19-positive LEVs were observed in diagnostic samples and isolated from mice engrafted with BCP-ALL primary cells. LEVs exhibited dynamic shape change in vitro and were internalised by other leukaemic cell lines leading to phenotypic transformation analogous to the cell of origin. In patient-derived xenografts, LEVs were released by primary ALL cells into extracellular spaces and internalised by murine mesenchymal cells in vivo. Collectively these data highlight the heterogeneity but accessibility of LEVs in clinical samples and their potential to provide a unique insight into the biology of the cell of origin and to their development as novel biomarkers to aid diagnosis and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Suzanne M Johnson
- Children's Cancer Group, Division of Molecular and Clinical Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester , Manchester , UK
| | - Clare Dempsey
- Children's Cancer Group, Division of Molecular and Clinical Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester , Manchester , UK
| | - Catriona Parker
- Children's Cancer Group, Division of Molecular and Clinical Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester , Manchester , UK
| | - Aleksandr Mironov
- Children's Cancer Group, Division of Molecular and Clinical Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester , Manchester , UK
| | - Helen Bradley
- Faculty of Biology, Medicine and Health, CRUK Manchester Institute, University of Manchester , Manchester , UK
| | - Vaskar Saha
- Children's Cancer Group, Division of Molecular and Clinical Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; Tata Translational Cancer Research Centre, Tata Medical Center, Kolkata, India
| |
Collapse
|
115
|
Rakobowchuk M, Ritter O, Wilhelm EN, Isacco L, Bouhaddi M, Degano B, Tordi N, Mourot L. Divergent endothelial function but similar platelet microvesicle responses following eccentric and concentric cycling at a similar aerobic power output. J Appl Physiol (1985) 2017; 122:1031-1039. [PMID: 28153942 DOI: 10.1152/japplphysiol.00602.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 01/03/2017] [Accepted: 01/27/2017] [Indexed: 01/06/2023] Open
Abstract
Endothelial function and microvesicle concentration changes after acute bouts of continuous eccentric exercise have not been assessed previously nor compared with concentric exercise at similar aerobic power outputs. This method of training may be useful among some clinical populations, but acute responses are not well described. As such, 12 healthy males completed 2 experimental sessions of either 45 min of eccentric or concentric cycling at a matched aerobic power output below the ventilatory threshold. Brachial artery vascular function was assessed throughout 5 min of forearm ischemia and 3 min thereafter, before and at 5 and 40 min of recovery following each exercise session [flow-mediated dilation (FMD)]. Venous blood samples were acquired before each vascular function assessment. FMD significantly decreased after eccentric cycling by 40 min of recovery (P < 0.05), but was unaltered after concentric exercise. No differences in peak hyperemic blood flow velocity occurred neither between modalities nor at any time point (P > 0.05). Platelet-derived microvesicles increased by ~20% after both exercise modalities (P < 0.05) while endothelial-derived microvesicles were unchanged (P > 0.05). Moderate relationships with cardiac output, a surrogate for shear stress, and norepinephrine were apparent (P < 0.05), but there were no relationships with inflammatory or acute phase proteins. In summary, eccentric endurance exercise induced macrovascular endothelial dysfunction; however, endothelial activation determined by endothelial microvesicles did not occur suggesting that this modality may induce oxidative stress but no significant endothelial damage. In addition, the increase in platelet microvesicle concentrations may induce beneficial microvascular adaptations as suggested by previous research.NEW & NOTEWORTHY Continuous eccentric cycling exercise induces substantial skeletal muscle, tendon, and bone strain providing a potentially beneficial stimulus among clinical populations. This modality also induces temporary endothelial dysfunction but no apparent damage or activation of the endothelium indicated by microvesicle production, whereas proangiogenic platelet microvesicles are released similarly following both concentric and eccentric cycling and may relate to the shear stress and catecholamine response to exercise.
Collapse
Affiliation(s)
- Mark Rakobowchuk
- Department of Biological Sciences, Faculty of Science, Thompson Rivers University, Kamloops, Canada;
| | - Ophélie Ritter
- EA 4267 Exercise Performance Health Innovation Platform, University Bourgogne Franche-Comté University, Besancon, France
| | - Eurico Nestor Wilhelm
- Centre for Sports Medicine and Human Performance, Brunel University London, London, United Kingdom; and
| | - Laurie Isacco
- EA 3920 Exercise Performance Health Innovation Platform, University Bourgogne Franche-Comté University, Besançon, France
| | - Malika Bouhaddi
- EA 3920 Exercise Performance Health Innovation Platform, University Bourgogne Franche-Comté University, Besançon, France
| | - Bruno Degano
- EA 3920 Exercise Performance Health Innovation Platform, University Bourgogne Franche-Comté University, Besançon, France
| | - Nicolas Tordi
- EA 4267 Exercise Performance Health Innovation Platform, University Bourgogne Franche-Comté University, Besancon, France
| | - Laurent Mourot
- EA 3920 Exercise Performance Health Innovation Platform, University Bourgogne Franche-Comté University, Besançon, France
| |
Collapse
|
116
|
Gajos K, Kamińska A, Awsiuk K, Bajor A, Gruszczyński K, Pawlak A, Żądło A, Kowalik A, Budkowski A, Stępień E. Immobilization and detection of platelet-derived extracellular vesicles on functionalized silicon substrate: cytometric and spectrometric approach. Anal Bioanal Chem 2017; 409:1109-1119. [PMID: 27822644 PMCID: PMC5258792 DOI: 10.1007/s00216-016-0036-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/28/2016] [Accepted: 10/17/2016] [Indexed: 12/21/2022]
Abstract
Among the various biomarkers that are used to diagnose or monitor disease, extracellular vesicles (EVs) represent one of the most promising targets in the development of new therapeutic strategies and the application of new diagnostic methods. The detection of circulating platelet-derived microvesicles (PMVs) is a considerable challenge for laboratory diagnostics, especially in the preliminary phase of a disease. In this study, we present a multistep approach to immobilizing and detecting PMVs in biological samples (microvesicles generated from activated platelets and human platelet-poor plasma) on functionalized silicon substrate. We describe the application of time-of-flight secondary ion mass spectrometry (TOF-SIMS) and spectroscopic ellipsometry methods to the detection of immobilized PMVs in the context of a novel imaging flow cytometry (ISX) technique and atomic force microscopy (AFM). This novel approach allowed us to confirm the presence of the abundant microvesicle phospholipids phosphatidylserine (PS) and phosphatidylethanolamine (PE) on a surface with immobilized PMVs. Phosphatidylcholine groups (C5H12N+; C5H15PNO4+) were also detected. Moreover, we were able to show that ellipsometry permitted the immobilization of PMVs on a functionalized surface to be evaluated. The sensitivity of the ISX technique depends on the size and refractive index of the analyzed microvesicles. Graphical abstract Human platelets activated with thrombin (in concentration 1IU/mL) generate population of PMVs (platelet derived microvesicles), which can be detected and enumerated with fluorescent-label method (imaging cytometry). Alternatively, PMVs can be immobilized on the modified silicon substrate which is functionalized with a specific IgM murine monoclonal antibody against human glycoprotein IIb/IIIa complex (PAC-1). Immobilized PMVs can be subjected to label-free analyses by means ellipsometry, atomic force microscopy (AFM) and time-of-flight secondary ion mass spectrometry (TOF-SIMS).
Collapse
Affiliation(s)
- Katarzyna Gajos
- Department of Advanced Materials Engineering, M. Smoluchowski Institute of Physics, Jagiellonian University, 11 Łojasiewicza Street, 30-348, Krakow, Poland
| | - Agnieszka Kamińska
- Department of Medical Physics, M. Smoluchowski Institute of Physics, Jagiellonian University, ul. S. Łojasiewicza 11, 30-348, Krakow, Poland
| | - Kamil Awsiuk
- Department of Advanced Materials Engineering, M. Smoluchowski Institute of Physics, Jagiellonian University, 11 Łojasiewicza Street, 30-348, Krakow, Poland
| | - Adrianna Bajor
- Department of Advanced Materials Engineering, M. Smoluchowski Institute of Physics, Jagiellonian University, 11 Łojasiewicza Street, 30-348, Krakow, Poland
| | - Krzysztof Gruszczyński
- Department of Molecular Diagnostics, Holycross Cancer Center, 3 Stefana Artwińskiego Street, 25-734, Kielce, Poland
| | - Anna Pawlak
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa Street, 30-387, Krakow, Poland
| | - Andrzej Żądło
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa Street, 30-387, Krakow, Poland
| | - Artur Kowalik
- Department of Molecular Diagnostics, Holycross Cancer Center, 3 Stefana Artwińskiego Street, 25-734, Kielce, Poland
| | - Andrzej Budkowski
- Department of Advanced Materials Engineering, M. Smoluchowski Institute of Physics, Jagiellonian University, 11 Łojasiewicza Street, 30-348, Krakow, Poland
| | - Ewa Stępień
- Department of Medical Physics, M. Smoluchowski Institute of Physics, Jagiellonian University, ul. S. Łojasiewicza 11, 30-348, Krakow, Poland.
| |
Collapse
|
117
|
Roussel C, Dussiot M, Marin M, Morel A, Ndour PA, Duez J, Le Van Kim C, Hermine O, Colin Y, Buffet PA, Amireault P. Spherocytic shift of red blood cells during storage provides a quantitative whole cell-based marker of the storage lesion. Transfusion 2017; 57:1007-1018. [PMID: 28150311 DOI: 10.1111/trf.14015] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/30/2016] [Accepted: 11/28/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Storage lesion may explain the rapid clearance of up to 25% of transfused red blood cells (RBCs) in recipients. Several alterations affect stored RBC but a quantitative, whole cell-based predictor of transfusion yield is lacking. Because RBCs with reduced surface area are retained by the spleen, we quantified changes in RBC dimensions during storage. STUDY DESIGN AND METHODS Using imaging flow cytometry we observed the dimension and morphology of RBCs upon storage, along with that of conventional biochemical and mechanical markers of storage lesion. We then validated these findings using differential interference contrast (DIC) microscopy and quantified the accumulation of microparticles (MPs). RESULTS Mean projected surface area of the whole RBC population decreased from 72.4 to 68.4 µm2 , a change resulting from the appearance of a well-demarcated subpopulation of RBCs with reduced mean projected surface (58 µm2 , 15.2%-19.9% reduction). These "small RBCs" accounted for 4.9 and 23.6% of all RBCs on Days 3 and 42 of storage, respectively. DIC microscopy confirmed that small RBCs had shifted upon storage from discocytes to echinocytes III, spheroechinocytes, and spherocytes. Glycophorin A-positive MPs and small RBCs appeared after similar kinetics. CONCLUSION The reduction in surface area of small RBCs is expected to induce their retention by the spleen. We propose that small RBCs generated by MP-induced membrane loss are preferentially cleared from the circulation shortly after transfusion of long-stored blood. Their operator-independent quantification using imaging flow cytometry may provide a marker of storage lesion potentially predictive of transfusion yield.
Collapse
Affiliation(s)
- Camille Roussel
- Université Sorbonne Paris Cité, Université Paris Descartes, Inserm, INTS, Unité Biologie Intégrée du Globule Rouge.,Université Sorbonne Paris Cité, Université Paris Descartes, Inserm, CNRS, Institut Imagine, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, Laboratoire d'Excellence GR-Ex
| | - Michaël Dussiot
- Université Sorbonne Paris Cité, Université Paris Descartes, Inserm, CNRS, Institut Imagine, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, Laboratoire d'Excellence GR-Ex
| | - Mickaël Marin
- Université Sorbonne Paris Cité, Université Paris Diderot, Inserm, INTS, Unité Biologie Intégrée du Globule Rouge
| | - Alexandre Morel
- Université Sorbonne Paris Cité, Université Paris Descartes, Inserm, CNRS, Institut Imagine, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, Laboratoire d'Excellence GR-Ex
| | - Papa Alioune Ndour
- Université Sorbonne Paris Cité, Université Paris Descartes, Inserm, INTS, Unité Biologie Intégrée du Globule Rouge
| | - Julien Duez
- Université Sorbonne Paris Cité, Université Paris Descartes, Inserm, INTS, Unité Biologie Intégrée du Globule Rouge
| | - Caroline Le Van Kim
- Université Sorbonne Paris Cité, Université Paris Diderot, Inserm, INTS, Unité Biologie Intégrée du Globule Rouge
| | - Olivier Hermine
- Université Sorbonne Paris Cité, Université Paris Descartes, Inserm, CNRS, Institut Imagine, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, Laboratoire d'Excellence GR-Ex.,Department of Adult Hematology, Necker Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Yves Colin
- Université Sorbonne Paris Cité, Université Paris Diderot, Inserm, INTS, Unité Biologie Intégrée du Globule Rouge
| | - Pierre A Buffet
- Université Sorbonne Paris Cité, Université Paris Descartes, Inserm, INTS, Unité Biologie Intégrée du Globule Rouge
| | - Pascal Amireault
- Université Sorbonne Paris Cité, Université Paris Diderot, Inserm, INTS, Unité Biologie Intégrée du Globule Rouge.,Université Sorbonne Paris Cité, Université Paris Descartes, Inserm, CNRS, Institut Imagine, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, Laboratoire d'Excellence GR-Ex
| |
Collapse
|
118
|
Camacho V, Toxavidis V, Tigges JC. Characterization of Extracellular Vesicles by Flow Cytometry. Methods Mol Biol 2017; 1660:175-190. [PMID: 28828656 DOI: 10.1007/978-1-4939-7253-1_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Here, we describe a comprehensive methodology for the setup and standardization of EV analysis using nanoscale flow cytometry. Controls of different size ranges, fluorescent intensities, and materials can be used to set up distribution curves that are then used for instrument optimization and as a reference guide. Using these controls, flow cytometry instruments can be primed for the detection, analysis, and sorting of specific EV populations. This allows for cross platform comparison and the ability to monitor both quality control (QC) and quality assurance (QA). The method here describes the use of nanoparticles to optimize a flow cytometer for small particle detection. It also outlines the procedures necessary to recover EVs for downstream applications.
Collapse
Affiliation(s)
- Virginia Camacho
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Vasilis Toxavidis
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - John C Tigges
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA. .,Flow Cytometry Core, Beth Israel Deaconess Medical Center, CLS-0932, 330 Brookline Ave., Boston, MA, 02215, USA.
| |
Collapse
|
119
|
Lukacs-Kornek V, Julich-Haertel H, Urban SK, Kornek M. Multi-Surface Antigen Staining of Larger Extracellular Vesicles. Methods Mol Biol 2017; 1660:201-208. [PMID: 28828658 DOI: 10.1007/978-1-4939-7253-1_16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Larger extracellular vesicles, microparticles (MPs) or microvesicles (MVs), especially their acquisition and characterization by flow cytometry (FACS), is increasingly in focus of clinical/translational research efforts. Several laboratories have shown that MPs/MVs might be suitable for the diagnosis and predicting prognosis in various diseases including cancer. However, FACS staining of larger extracellular vesicles (EVs) can be difficult and results potentially in false positive and inconsistent data interpretation. Despite that FACS equipment is well maintained and the operators have ample experience, a reliable and for larger EVs optimized staining protocol is missing. Here, we aim to close that gap and provide a working multi-antibody staining protocol for larger EVs isolated from human serum samples. We describe in detail the needed steps as currently done in our laboratory. Staining is demonstrated exemplarily for multi-antibody mix including CD147 , a potential cancer marker if applied in combination with other MP/MV surface markers.
Collapse
Affiliation(s)
- Veronika Lukacs-Kornek
- Department of Medicine II, Saarland University Medical Center, Saarland University, Kirrbergerstr. 100, Homburg, 66424, Germany
| | - Henrike Julich-Haertel
- Department of Medicine II, Saarland University Medical Center, Saarland University, Kirrbergerstr. 100, Homburg, 66424, Germany
| | - Sabine Katharina Urban
- Department of Medicine II, Saarland University Medical Center, Saarland University, Kirrbergerstr. 100, Homburg, 66424, Germany
| | - Miroslaw Kornek
- Department of Medicine II, Saarland University Medical Center, Saarland University, Kirrbergerstr. 100, Homburg, 66424, Germany.
| |
Collapse
|
120
|
Haridas V, Ranjbar S, Vorobjev IA, Goldfeld AE, Barteneva NS. Imaging flow cytometry analysis of intracellular pathogens. Methods 2017; 112:91-104. [PMID: 27642004 PMCID: PMC5857943 DOI: 10.1016/j.ymeth.2016.09.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 08/15/2016] [Accepted: 09/15/2016] [Indexed: 01/09/2023] Open
Abstract
Imaging flow cytometry has been applied to address questions in infection biology, in particular, infections induced by intracellular pathogens. This methodology, which utilizes specialized analytic software makes it possible to analyze hundreds of quantified features for hundreds of thousands of individual cellular or subcellular events in a single experiment. Imaging flow cytometry analysis of host cell-pathogen interaction can thus quantitatively addresses a variety of biological questions related to intracellular infection, including cell counting, internalization score, and subcellular patterns of co-localization. Here, we provide an overview of recent achievements in the use of fluorescently labeled prokaryotic or eukaryotic pathogens in human cellular infections in analysis of host-pathogen interactions. Specifically, we give examples of Imagestream-based analysis of cell lines infected with Toxoplasma gondii or Mycobacterium tuberculosis. Furthermore, we illustrate the capabilities of imaging flow cytometry using a combination of standard IDEAS™ software and the more recently developed Feature Finder algorithm, which is capable of identifying statistically significant differences between researcher-defined image galleries. We argue that the combination of imaging flow cytometry with these software platforms provides a powerful new approach to understanding host control of intracellular pathogens.
Collapse
Affiliation(s)
- Viraga Haridas
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, United States; Department of Pediatrics, Harvard Medical School, United States
| | - Shahin Ranjbar
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, United States; Department of Pediatrics, Harvard Medical School, United States
| | - Ivan A Vorobjev
- School of Science and Technology, Nazarbayev University, Kazakhstan; A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Russia; Department of Cell Biology and Histology, M.V. Lomonosov Moscow State University, Russia
| | - Anne E Goldfeld
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, United States; Department of Pediatrics, Harvard Medical School, United States.
| | - Natasha S Barteneva
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, United States; Department of Pediatrics, Harvard Medical School, United States; School of Science and Technology, Nazarbayev University, Kazakhstan.
| |
Collapse
|
121
|
Gasecka A, Böing AN, Filipiak KJ, Nieuwland R. Platelet extracellular vesicles as biomarkers for arterial thrombosis. Platelets 2016; 28:228-234. [PMID: 27996341 DOI: 10.1080/09537104.2016.1254174] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Arterial thrombosis is a major and global cause of human death and disability. Considering the socioeconomic costs of arterial thrombosis, identification of biomarkers to predict and detect arterial thrombosis at an early stage is an important public health goal. Platelet extracellular vesicles (PEV) are a new candidate biomarker of arterial thrombosis. PEV can be measured in biorepositories, thereby offering the possibility to validate PEV in multicenter clinical trials. PEV analysis has been hitherto hampered by lack of standardized methodology, but substantial technological improvements of PEV detection techniques have been achieved recently. However, before PEV emerge from research tools to clinical applications, a number of issues should be clarified. To facilitate validation of PEV as biomarkers of thrombosis, we discuss (i) whether PEV are useful as biomarkers of thrombosis, (ii) why previous conclusions on PEV concentrations, composition and functions require re-evaluation, and (iii) which questions have to be answered before PEV become clinically useful.
Collapse
Affiliation(s)
- Aleksandra Gasecka
- a 1st Chair and Department of Cardiology , Medical University of Warsaw , Warsaw , Poland.,b Laboratory of Experimental Clinical Chemistry, and Vesicle Observation Centre , Academic Medical Centre of the University of Amsterdam , Amsterdam , The Netherlands
| | - Anita N Böing
- b Laboratory of Experimental Clinical Chemistry, and Vesicle Observation Centre , Academic Medical Centre of the University of Amsterdam , Amsterdam , The Netherlands
| | - Krzysztof J Filipiak
- a 1st Chair and Department of Cardiology , Medical University of Warsaw , Warsaw , Poland
| | - Rienk Nieuwland
- b Laboratory of Experimental Clinical Chemistry, and Vesicle Observation Centre , Academic Medical Centre of the University of Amsterdam , Amsterdam , The Netherlands
| |
Collapse
|
122
|
Acker JP, Marks DC, Sheffield WP. Quality Assessment of Established and Emerging Blood Components for Transfusion. JOURNAL OF BLOOD TRANSFUSION 2016; 2016:4860284. [PMID: 28070448 PMCID: PMC5192317 DOI: 10.1155/2016/4860284] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/02/2016] [Indexed: 12/16/2022]
Abstract
Blood is donated either as whole blood, with subsequent component processing, or through the use of apheresis devices that extract one or more components and return the rest of the donation to the donor. Blood component therapy supplanted whole blood transfusion in industrialized countries in the middle of the twentieth century and remains the standard of care for the majority of patients receiving a transfusion. Traditionally, blood has been processed into three main blood products: red blood cell concentrates; platelet concentrates; and transfusable plasma. Ensuring that these products are of high quality and that they deliver their intended benefits to patients throughout their shelf-life is a complex task. Further complexity has been added with the development of products stored under nonstandard conditions or subjected to additional manufacturing steps (e.g., cryopreserved platelets, irradiated red cells, and lyophilized plasma). Here we review established and emerging methodologies for assessing blood product quality and address controversies and uncertainties in this thriving and active field of investigation.
Collapse
Affiliation(s)
- Jason P. Acker
- Centre for Innovation, Canadian Blood Services, Edmonton, AB, Canada
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Denese C. Marks
- Research and Development, Australian Red Cross Blood Service, Sydney, NSW, Australia
| | - William P. Sheffield
- Centre for Innovation, Canadian Blood Services, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
123
|
Bhullar J, Bhopale VM, Yang M, Sethuraman K, Thom SR. Microparticle formation by platelets exposed to high gas pressures - An oxidative stress response. Free Radic Biol Med 2016; 101:154-162. [PMID: 27751909 DOI: 10.1016/j.freeradbiomed.2016.10.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/10/2016] [Accepted: 10/13/2016] [Indexed: 02/05/2023]
Abstract
This investigation explored the mechanism for microparticles (MPs) production by human and murine platelets exposed to high pressures of inert gases. Results demonstrate that MPs production occurs via an oxidative stress response in a dose-dependent manner and follows the potency series N2>Ar>He. Gases with higher van der Waals volumes or polarizability such as SF6 and N2O, or hydrostatic pressure, do not cause MPs production. Singlet O2 is generated by N2, Ar and He, which is linked to NADPH oxidase (NOX) activity. Progression of oxidative stress involves activation of nitric oxide synthase (NOS) leading to S-nitrosylation of cytosolic actin. Exposure to gases enhances actin filament turnover and associations between short actin filaments, NOS, vasodilator-stimulated phosphoprotein (VASP), focal adhesion kinase (FAK) and Rac1. Inhibition of NOS or NOX by chemical inhibitors or using platelets from mice lacking NOS2 or the gp91phox component of NOX diminish generation of reactive species, enhanced actin polymerization and MP generation by high pressure gases. We conclude that by initiating a sequence of progressive oxidative stress responses high pressure gases cause platelets to generate MPs.
Collapse
Affiliation(s)
- Jasjeet Bhullar
- Department of Emergency Medicine, University of Maryland, Baltimore, MD 21201, United States
| | - Veena M Bhopale
- Department of Emergency Medicine, University of Maryland, Baltimore, MD 21201, United States
| | - Ming Yang
- Department of Emergency Medicine, University of Maryland, Baltimore, MD 21201, United States
| | - Kinjal Sethuraman
- Department of Emergency Medicine, University of Maryland, Baltimore, MD 21201, United States
| | - Stephen R Thom
- Department of Emergency Medicine, University of Maryland, Baltimore, MD 21201, United States.
| |
Collapse
|
124
|
Han Y, Gu Y, Zhang AC, Lo YH. Review: imaging technologies for flow cytometry. LAB ON A CHIP 2016; 16:4639-4647. [PMID: 27830849 PMCID: PMC5311077 DOI: 10.1039/c6lc01063f] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
High-throughput single cell imaging is a critical enabling and driving technology in molecular and cellular biology, biotechnology, medicine and related areas. Imaging flow cytometry combines the single-cell imaging capabilities of microscopy with the high-throughput capabilities of conventional flow cytometry. Recent advances in imaging flow cytometry are remarkably revolutionizing single-cell analysis. This article describes recent imaging flow cytometry technologies and their challenges.
Collapse
Affiliation(s)
- Yuanyuan Han
- Department of Electrical and Computer Engineering, University of California, San Diego, California 92093, USA.
| | - Yi Gu
- Department of Electrical and Computer Engineering, University of California, San Diego, California 92093, USA.
| | - Alex Ce Zhang
- Department of Electrical and Computer Engineering, University of California, San Diego, California 92093, USA.
| | - Yu-Hwa Lo
- Department of Electrical and Computer Engineering, University of California, San Diego, California 92093, USA.
| |
Collapse
|
125
|
Xu R, Fitts A, Li X, Fernandes J, Pochampally R, Mao J, Liu YM. Quantification of Small Extracellular Vesicles by Size Exclusion Chromatography with Fluorescence Detection. Anal Chem 2016; 88:10390-10394. [PMID: 27689436 PMCID: PMC5089915 DOI: 10.1021/acs.analchem.6b03348] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Chemical analysis of small extracellular vesicles (sEVs) circulating in body fluids holds potentials in noninvasive diagnosis of diseases and evaluation of therapeutic treatments. However, quantification of sEVs remains a challenge due to lacking of cost-effective analytical protocols. Herein we report a facile method based on size exclusion chromatography with fluorescence detection (SEC-FD) for sEVs quantification. After removal of cells and cell debris, a 0.50 mL sample (e.g., cell culture medium) is incubated with CM-Dil dye to fluorescently label sEVs. The incubation solution is then separated on a SEC column packed with Sepharose CL-4B. The eluent is monitored fluorescently at Ex553 nm/Em570 nm by using a fluorometer equipped with a 50-μL flow through cuvette. Separation efficiency of the proposed SEC-FD method was evaluated by analyzing 100 nm liposomes and albumin-FITC conjugate. Liposomes were eluted out in less than 6 min, about 10 min before albumin-FITC. A separation repeatability (RSD in retention time) of 1.4% (n = 5) was obtained for liposomes. In analysis of cell culture media, linear calibration curves based on SEC-FD peak height versus sEVs concentration were obtained with r2 value of 0.996. Intraday quantification repeatability (RSD in peak height) was 3.2% (n = 5). The detection limit was estimated to be 2.9 × 107 exosome particles/mL. The proposed assay was applied to the first study of sEVs secretion from TK6 cells cultured in serum-free medium for a culturing period from 1 to 48 h.
Collapse
Affiliation(s)
- Rui Xu
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, MS 39217, USA
| | - Austin Fitts
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, MS 39217, USA
| | - Xiangtang Li
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, MS 39217, USA
| | - Joseph Fernandes
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39150, USA
| | - Radhika Pochampally
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39150, USA
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS 39150, USA
| | - Jinghe Mao
- Department of Biology, Tougaloo College, Tougaloo, MS 39174, USA
| | - Yi-Ming Liu
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, MS 39217, USA
| |
Collapse
|
126
|
Lannigan J, Erdbruegger U. Imaging flow cytometry for the characterization of extracellular vesicles. Methods 2016; 112:55-67. [PMID: 27721015 DOI: 10.1016/j.ymeth.2016.09.018] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/15/2016] [Accepted: 09/30/2016] [Indexed: 12/21/2022] Open
Abstract
Extracellular Vesicles (EVs) are potent bio-activators and inter-cellular communicators that play an important role in both health and disease. It is for this reason there is a strong interest in understanding their composition and origin, with the hope of using them as important biomarkers or therapeutics. Due to their very small size, heterogeneity, and large numbers there has been a need for better tools to measure them in an accurate and high throughput manner. While traditional flow cytometry has been widely used for this purpose, there are inherent problems with this approach, as these instruments have traditionally been developed to measure whole cells, which are orders of magnitude larger and express many more molecules of identifying epitopes. Imaging flow cytometry, as performed with the ImagestreamX MKII, with its combination of increased fluorescence sensitivity, low background, image confirmation ability and powerful data analysis tools, provides a great tool to accurately evaluate EVs. We present here a comprehensive approach in applying this technology to the study of EVs.
Collapse
Affiliation(s)
- Joanne Lannigan
- University of Virginia, School of Medicine, Flow Cytometry Core, 1300 Jefferson Park Avenue, Charlottesville, VA 22908-0734, USA.
| | - Uta Erdbruegger
- University of Virginia, Department of Medicine/Nephrology Division, 1300 Jefferson Park Avenue, Charlottesville, VA 22908-0133, USA.
| |
Collapse
|
127
|
Eichholz K, Bru T, Tran TTP, Fernandes P, Welles H, Mennechet FJD, Manel N, Alves P, Perreau M, Kremer EJ. Immune-Complexed Adenovirus Induce AIM2-Mediated Pyroptosis in Human Dendritic Cells. PLoS Pathog 2016; 12:e1005871. [PMID: 27636895 PMCID: PMC5026364 DOI: 10.1371/journal.ppat.1005871] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 08/15/2016] [Indexed: 02/07/2023] Open
Abstract
Human adenoviruses (HAdVs) are nonenveloped proteinaceous particles containing a linear double-stranded DNA genome. HAdVs cause a spectrum of pathologies in all populations regardless of health standards. Following repeat exposure to multiple HAdV types, we develop robust and long-lived humoral and cellular immune responses that provide life-long protection from de novo infections and persistent HAdV. How HAdVs, anti-HAdV antibodies and antigen presenting cells (APCs) interact to influence infection is still incompletely understood. In our study, we used physical, pharmacological, biochemical, fluorescence and electron microscopy, molecular and cell biology approaches to dissect the impact of immune-complexed HAdV (IC-HAdV) on human monocyte-derived dendritic cells (MoDCs). We show that IC-HAdV generate stabilized complexes of ~200 nm that are efficiently internalized by, and aggregate in, MoDCs. By comparing IC-HAdV, IC-empty capsid, IC-Ad2ts1 (a HAdV-C2 impaired in endosomal escape due to a mutation that impacts protease encapsidation) and IC-AdL40Q (a HAdV-C5 impaired in endosomal escape due to a mutation in protein VI), we demonstrate that protein VI-dependent endosomal escape is required for the HAdV genome to engage the DNA pattern recognition receptor AIM2 (absent in melanoma 2). AIM2 engagement induces pyroptotic MoDC death via ASC (apoptosis-associated speck protein containing a caspase activation/recruitment domain) aggregation, inflammasome formation, caspase 1 activation, and IL-1β and gasdermin D (GSDMD) cleavage. Our study provides mechanistic insight into how humoral immunity initiates an innate immune response to HAdV-C5 in human professional APCs.
Collapse
Affiliation(s)
- Karsten Eichholz
- Institut de Génétique Moléculaire de Montpellier, CNRS 5535, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - Thierry Bru
- Institut de Génétique Moléculaire de Montpellier, CNRS 5535, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - Thi Thu Phuong Tran
- Institut de Génétique Moléculaire de Montpellier, CNRS 5535, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - Paulo Fernandes
- iBET- Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Hugh Welles
- Division of Immunology and Allergy, University of Lausanne, Lausanne, Switzerland
| | - Franck J. D. Mennechet
- Institut de Génétique Moléculaire de Montpellier, CNRS 5535, Montpellier, France
- Université de Montpellier, Montpellier, France
| | | | - Paula Alves
- iBET- Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Matthieu Perreau
- Division of Immunology and Allergy, University of Lausanne, Lausanne, Switzerland
| | - Eric J. Kremer
- Institut de Génétique Moléculaire de Montpellier, CNRS 5535, Montpellier, France
- Université de Montpellier, Montpellier, France
- * E-mail:
| |
Collapse
|
128
|
Wilhelm EN, González-Alonso J, Parris C, Rakobowchuk M. Exercise intensity modulates the appearance of circulating microvesicles with proangiogenic potential upon endothelial cells. Am J Physiol Heart Circ Physiol 2016; 311:H1297-H1310. [PMID: 27638881 DOI: 10.1152/ajpheart.00516.2016] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 09/12/2016] [Indexed: 12/22/2022]
Abstract
The effect of endurance exercise on circulating microvesicle dynamics and their impact on surrounding endothelial cells is unclear. Here we tested the hypothesis that exercise intensity modulates the time course of platelet (PMV) and endothelial-derived (EMV) microvesicle appearance in the circulation through hemodynamic and biochemical-related mechanisms, and that microvesicles formed during exercise would stimulate endothelial angiogenesis in vitro. Nine healthy young men had venous blood samples taken before, during, and throughout the recovery period after 1 h of moderate [46 ± 2% maximal oxygen uptake (V̇o2max)] or heavy (67 ± 2% V̇o2max) intensity semirecumbent cycling and a time-matched resting control trial. In vitro experiments were performed by incubating endothelial cells with rest and exercise-derived microvesicles to examine their effects on cell angiogenic capacities. PMVs (CD41+) increased from baseline only during heavy exercise (from 21 ± 1 × 103 to 55 ± 8 × 103 and 48 ± 6 × 103 PMV/μl at 30 and 60 min, respectively; P < 0.05), returning to baseline early in postexercise recovery (P > 0.05), whereas EMVs (CD62E+) were unchanged (P > 0.05). PMVs were related to brachial artery shear rate (r2 = 0.43) and plasma norepinephrine concentrations (r2 = 0.21) during exercise (P < 0.05). Exercise-derived microvesicles enhanced endothelial proliferation, migration, and tubule formation compared with rest microvesicles (P < 0.05). These results demonstrate substantial increases in circulating PMVs during heavy exercise and that exercise-derived microvesicles stimulate human endothelial cells by enhancing angiogenesis and proliferation. This involvement of microvesicles may be considered a novel mechanism through which exercise mediates vascular healing and adaptation.
Collapse
Affiliation(s)
- Eurico N Wilhelm
- Centre for Human Performance, Exercise and Rehabilitation, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - José González-Alonso
- Centre for Human Performance, Exercise and Rehabilitation, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Christopher Parris
- Institute for the Environment, Health and Societies, Brunel University London, Uxbridge, United Kingdom; and
| | - Mark Rakobowchuk
- Faculty of Science, Department of Biological Sciences, Thompson Rivers University Kamloops, British Columbia, Canada
| |
Collapse
|
129
|
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
|
130
|
Cesi G, Walbrecq G, Margue C, Kreis S. Transferring intercellular signals and traits between cancer cells: extracellular vesicles as "homing pigeons". Cell Commun Signal 2016; 14:13. [PMID: 27282631 PMCID: PMC4901437 DOI: 10.1186/s12964-016-0136-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/02/2016] [Indexed: 12/15/2022] Open
Abstract
Extracellular vesicles are cell-derived vesicles, which can transport various cargos out of cells. From their cell of origin, the content molecules (proteins, non-coding RNAs including miRNAs, DNA and others) can be delivered to neighboring or distant cells and as such extracellular vesicles can be regarded as vehicles of intercellular communication or "homing pigeons". Extracellular vesicle shuttling is able to actively modulate the tumor microenvironment and can partake in tumor dissemination. In various diseases, including cancer, levels of extracellular vesicle secretion are altered resulting in different amounts and/or profiles of detectable vesicular cargo molecules and these distinct content profiles are currently being evaluated as biomarkers. Apart from their potential as blood-derived containers of specific biomarkers, the transfer of extracellular vesicles to surrounding cells also appears to be involved in the propagation of phenotypic traits. These interesting properties have put extracellular vesicles into the focus of many recent studies.Here we review findings on the involvement of extracellular vesicles in transferring traits of cancer cells to their surroundings and briefly discuss new data on oncosomes, a larger type of vesicle. A pressing issue in cancer treatment is rapidly evolving resistance to many initially efficient drug therapies. Studies investigating the role of extracellular vesicles in this phenomenon together with a summary of the technical challenges that this field is still facing, are also presented. Finally, emerging areas of research such as the analysis of the lipid composition on extracellular vesicles and cutting-edge techniques to visualise the trafficking of extracellular vesicles are discussed.
Collapse
Affiliation(s)
- Giulia Cesi
- Life Sciences Research Unit, University of Luxembourg, 6, av. du Swing, L-4367, Belvaux, Luxembourg
| | - Geoffroy Walbrecq
- Life Sciences Research Unit, University of Luxembourg, 6, av. du Swing, L-4367, Belvaux, Luxembourg
| | - Christiane Margue
- Life Sciences Research Unit, University of Luxembourg, 6, av. du Swing, L-4367, Belvaux, Luxembourg
| | - Stephanie Kreis
- Life Sciences Research Unit, University of Luxembourg, 6, av. du Swing, L-4367, Belvaux, Luxembourg.
| |
Collapse
|
131
|
Augustine TN, van der Spuy WJ, Kaberry LL, Shayi M. Thrombin-Mediated Platelet Activation of Lysed Whole Blood and Platelet-Rich Plasma: A Comparison Between Platelet Activation Markers and Ultrastructural Alterations. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2016; 22:630-639. [PMID: 27329313 DOI: 10.1017/s1431927616000854] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Platelet ultrastructural alterations representing spurious activation have been identified in pathological conditions. A limitation of platelet studies is that sample preparation may lead to artifactual activation processes which may confound results, impacting the use of scanning electron microscopy as a supplemental diagnostic tool. We used scanning electron microscopy and flow cytometry to analyze platelet activation in platelet-rich plasma (PRP) and whole blood (WB) samples. PRP generated using a single high g force centrifugation, and WB samples treated with a red blood cell lysis buffer, were exposed to increasing concentrations of the agonist thrombin. Platelets in lysed WB samples responded to thrombin by elevating the activation marker CD62p definitively, with corresponding ultrastructural changes indicating activation. Conversely, CD62p expression in PRP preparations remained static. Ultrastructural analysis revealed fully activated platelets even under low concentration thrombin stimulation, with considerable fibrin deposition. It is proposed that the method for PRP production induced premature platelet activation, preventable by using an inhibitor of platelet aggregation and fibrin polymerization. Nevertheless, our results show a definitive correspondence between flow cytometry and scanning electron microscopy in platelet activation studies, highlighting the potential of the latter technique as a supplemental diagnostic tool.
Collapse
Affiliation(s)
- Tanya N Augustine
- School of Anatomical Sciences, Faculty of Health Sciences,University of the Witwatersrand,Johannesburg,South Africa
| | - Wendy J van der Spuy
- School of Anatomical Sciences, Faculty of Health Sciences,University of the Witwatersrand,Johannesburg,South Africa
| | - Lindsay L Kaberry
- School of Anatomical Sciences, Faculty of Health Sciences,University of the Witwatersrand,Johannesburg,South Africa
| | - Millicent Shayi
- School of Anatomical Sciences, Faculty of Health Sciences,University of the Witwatersrand,Johannesburg,South Africa
| |
Collapse
|
132
|
Neri T, Lombardi S, Faìta F, Petrini S, Balìa C, Scalise V, Pedrinelli R, Paggiaro P, Celi A. Pirfenidone inhibits p38-mediated generation of procoagulant microparticles by human alveolar epithelial cells. Pulm Pharmacol Ther 2016; 39:1-6. [PMID: 27237042 DOI: 10.1016/j.pupt.2016.05.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 05/23/2016] [Accepted: 05/25/2016] [Indexed: 02/01/2023]
Abstract
Pirfenidone is a drug recently approved for idiopathic pulmonary fibrosis but its mechanisms of action are partially unknown. We have previously demonstrated that the airways of patients with idiopathic pulmonary fibrosis contain procoagulant microparticles that activate coagulation factor X to its active form, Xa, a proteinase that signals fibroblast growth and differentiation, thus potentially contributing to the pathogenesis of the disease. We also reported that in vitro exposure of human alveolar cells to H2O2 causes microparticle generation. Since p38 activation is involved in microparticle generation in some cell models and p38 inhibition is one of the mechanisms of action of pirfenidone, we investigated the hypothesis that H2O2-induced generation of microparticles by alveolar cells is dependent on p38 phosphorylation and is inhibited by pirfenidone. H2O2 stimulation of alveolar cells caused p38 phosphorylation that was inhibited by pirfenidone. The drug also inhibited H2O2 induced microparticle generation as assessed by two independent methods (solid phase thrombin generation and flow cytometry). The shedding of microparticle-bound tissue factor activity was also inhibited by pirfenidone. Inhibition of p38-mediated generation of procoagulant microparticle is a previously unrecognized mechanism of action of the antifibrotic drug, pirfenidone.
Collapse
Affiliation(s)
- Tommaso Neri
- Laboratorio di Biologia Cellulare Respiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e di Area Critica, Università di Pisa, Pisa, Italy
| | - Stefania Lombardi
- SSD Analisi ChimicoCliniche ed ImmunoAllergologia, USL1, Massa e Carrara, Italy
| | - Francesca Faìta
- Laboratorio di Biologia Cellulare Respiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e di Area Critica, Università di Pisa, Pisa, Italy
| | - Silvia Petrini
- Laboratorio di Biologia Cellulare Respiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e di Area Critica, Università di Pisa, Pisa, Italy
| | - Cristina Balìa
- Laboratorio di Biologia Cellulare Respiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e di Area Critica, Università di Pisa, Pisa, Italy
| | - Valentina Scalise
- Laboratorio di Biologia Cellulare Respiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e di Area Critica, Università di Pisa, Pisa, Italy
| | - Roberto Pedrinelli
- Laboratorio di Biologia Cellulare Respiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e di Area Critica, Università di Pisa, Pisa, Italy
| | - Pierluigi Paggiaro
- Laboratorio di Biologia Cellulare Respiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e di Area Critica, Università di Pisa, Pisa, Italy
| | - Alessandro Celi
- Laboratorio di Biologia Cellulare Respiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e di Area Critica, Università di Pisa, Pisa, Italy.
| |
Collapse
|
133
|
The role of neutrophils in inflammation resolution. Semin Immunol 2016; 28:137-45. [DOI: 10.1016/j.smim.2016.03.007] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 03/11/2016] [Accepted: 03/15/2016] [Indexed: 01/29/2023]
|
134
|
Neri T, Pergoli L, Petrini S, Gravendonk L, Balia C, Scalise V, Amoruso A, Pedrinelli R, Paggiaro P, Bollati V, Celi A. Particulate matter induces prothrombotic microparticle shedding by human mononuclear and endothelial cells. Toxicol In Vitro 2016; 32:333-8. [DOI: 10.1016/j.tiv.2016.02.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 02/06/2016] [Indexed: 12/31/2022]
|
135
|
Xu R, Greening DW, Zhu HJ, Takahashi N, Simpson RJ. Extracellular vesicle isolation and characterization: toward clinical application. J Clin Invest 2016; 126:1152-62. [PMID: 27035807 DOI: 10.1172/jci81129] [Citation(s) in RCA: 659] [Impact Index Per Article: 73.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Two broad categories of extracellular vesicles (EVs), exosomes and shed microvesicles (sMVs), which differ in size distribution as well as protein and RNA profiles, have been described. EVs are known to play key roles in cell-cell communication, acting proximally as well as systemically. This Review discusses the nature of EV subtypes, strategies for isolating EVs from both cell-culture media and body fluids, and procedures for quantifying EVs. We also discuss proteins selectively enriched in exosomes and sMVs that have the potential for use as markers to discriminate between EV subtypes, as well as various applications of EVs in clinical diagnosis.
Collapse
|
136
|
Yao D, Lin Z, Wu J. Near-Infrared Fluorogenic Probes with Polarity-Sensitive Emission for in Vivo Imaging of an Ovarian Cancer Biomarker. ACS APPLIED MATERIALS & INTERFACES 2016; 8:5847-5856. [PMID: 26910257 DOI: 10.1021/acsami.5b11826] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Lysophosphatidic acid (LPA, cutoff values ≥ 1.5 μM) is an effective biomarker for early stage ovarian cancer. The development of selective probes for LPA detection is therefore critical for early clinical diagnosis. Although current methods have been developed for the detection of LPA in solution, they cannot be used for tracking LPA in vivo. Here, we report a near-infrared (NIR) fluorescent probe that can selectively respond to LPA based on polarity-sensitive emission at a very low detection limit of 0.5 μM in situ. This probe exhibits a marked increase of fluorescence at 720 nm upon binding to LPA, allowing the direct visualization of LPA in vitro and in vivo without interference from other biomolecules. Moreover, the probe containing two arginine-glycine-aspartic acid units can be efficiently taken up by cancer cells based on an αvβ3 integrin receptor targeting mechanism. It also exhibits excellent biocompatibility and high pH stability in live cells and in vivo. Confocal laser scanning microscopy and flow cytometric imaging of SKOV-3 cells have confirmed that our probe can be used to image LPA in live cells. In particular, its NIR turn-on fluorescence can be used to effectively monitor LPA imaging in a SKOV-3 tumor-bearing mouse model. Our probe may pave the way for the detection of cancer-related biomarkers and even for early stage cancer diagnosis.
Collapse
Affiliation(s)
- Defan Yao
- Key Laboratory for Advanced Materials & Institute of Fine Chemicals, East China University of Science and Technology , Shanghai 200237, China
| | - Zhi Lin
- College of Chemistry and Molecular Engineering, Peking University , Beijing 100871, China
| | - Junchen Wu
- Key Laboratory for Advanced Materials & Institute of Fine Chemicals, East China University of Science and Technology , Shanghai 200237, China
| |
Collapse
|
137
|
Gao L, Wang LV. A review of snapshot multidimensional optical imaging: measuring photon tags in parallel. PHYSICS REPORTS 2016; 616:1-37. [PMID: 27134340 PMCID: PMC4846296 DOI: 10.1016/j.physrep.2015.12.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Multidimensional optical imaging has seen remarkable growth in the past decade. Rather than measuring only the two-dimensional spatial distribution of light, as in conventional photography, multidimensional optical imaging captures light in up to nine dimensions, providing unprecedented information about incident photons' spatial coordinates, emittance angles, wavelength, time, and polarization. Multidimensional optical imaging can be accomplished either by scanning or parallel acquisition. Compared with scanning-based imagers, parallel acquisition-also dubbed snapshot imaging-has a prominent advantage in maximizing optical throughput, particularly when measuring a datacube of high dimensions. Here, we first categorize snapshot multidimensional imagers based on their acquisition and image reconstruction strategies, then highlight the snapshot advantage in the context of optical throughput, and finally we discuss their state-of-the-art implementations and applications.
Collapse
Affiliation(s)
- Liang Gao
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, 306 N. Wright St., Urbana, Illinois 61801
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 North Mathews Avenue, Urbana, Illinois 61801
| | - Lihong V. Wang
- Optical imaging laboratory, Department of Biomedical Engineering, Washington University in St. Louis, One Brookings Dr., MO, 63130
| |
Collapse
|
138
|
Crawford JR, Trial J, Nambi V, Hoogeveen RC, Taffet GE, Entman ML. Plasma Levels of Endothelial Microparticles Bearing Monomeric C-reactive Protein are Increased in Peripheral Artery Disease. J Cardiovasc Transl Res 2016; 9:184-193. [PMID: 26891844 DOI: 10.1007/s12265-016-9678-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 01/22/2016] [Indexed: 01/24/2023]
Abstract
C-reactive protein (CRP) as an indicator of cardiovascular disease (CVD) has shown limited sensitivity. We demonstrate that two isoforms of CRP (pentameric, pCRP and monomeric, mCRP) present in soluble form or on microparticles (MPs) have different biological effects and are not all measured by clinical CRP assays. The high-sensitivity CRP assay (hsCRP) did not measure pCRP or mCRP on MPs, whereas flow cytometry did. MPs derived from endothelial cells, particularly those bearing mCRP, were elevated in peripheral artery disease (PAD) patients compared to controls. The numbers of mCRP(+) endothelial MPs did not correlate with hsCRP measurements of soluble pCRP, indicating their independent modulation. In controls, statins lowered mCRP(+) endothelial MPs. In a model of vascular inflammation, mCRP induced endothelial shedding of MPs and was proinflammatory, while pCRP was anti-inflammatory. mCRP on endothelial MPs may be both an unmeasured indicator of, and an amplifier of, vascular disease, and its detection might improve risk sensitivity.
Collapse
Affiliation(s)
- Jeffrey R Crawford
- The Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine and Houston Methodist Hospital, One Baylor Plaza, M.S. BCM620, Houston, TX, 77030, USA
| | - JoAnn Trial
- The Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine and Houston Methodist Hospital, One Baylor Plaza, M.S. BCM620, Houston, TX, 77030, USA.
| | - Vijay Nambi
- The Division of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.,The Division of Atherosclerosis and Vascular Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.,Center for Cardiovascular Prevention, Methodist DeBakey Heart and Vascular Center, 6565 Fannin St., Houston, TX, 77030, USA
| | - Ron C Hoogeveen
- The Division of Atherosclerosis and Vascular Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - George E Taffet
- The Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine and Houston Methodist Hospital, One Baylor Plaza, M.S. BCM620, Houston, TX, 77030, USA
| | - Mark L Entman
- The Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine and Houston Methodist Hospital, One Baylor Plaza, M.S. BCM620, Houston, TX, 77030, USA
| |
Collapse
|
139
|
Chandler WL. Measurement of microvesicle levels in human blood using flow cytometry. CYTOMETRY PART B-CLINICAL CYTOMETRY 2016; 90:326-36. [PMID: 26606416 DOI: 10.1002/cyto.b.21343] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 11/04/2015] [Accepted: 11/19/2015] [Indexed: 11/08/2022]
Abstract
Microvesicles are fragments of cells released when the cells are activated, injured, or apoptotic. Analysis of microvesicle levels in blood has the potential to shed new light on the pathophysiology of many diseases. Flow cytometry is currently the only method that can simultaneously separate true lipid microvesicles from other microparticles in blood, determine the cell of origin and other microvesicle characteristics, and handle large numbers of clinical samples with a reasonable effort, but expanded use of flow cytometric measurement of microvesicle levels as a clinical and research tool requires improved, standardized assays. The goal of this review is to aid investigators in applying current best practices to microvesicle measurements. First pre-analytical factors are evaluated and data summarized for anticoagulant effects, sample transport and centrifugation. Next flow cytometer optimization is reviewed including interference from background in buffers and reagents, accurate microvesicle counting, swarm interference, and other types of coincidence errors, size calibration, and detection limits using light scattering, impedance and fluorescence. Finally current progress on method standardization is discussed and a summary of current best practices provided. © 2016 Clinical Cytometry Society.
Collapse
Affiliation(s)
- Wayne L Chandler
- Department of Laboratories, Seattle Children's Hospital, and Department of Laboratory Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
140
|
Lawson C, Vicencio JM, Yellon DM, Davidson SM. Microvesicles and exosomes: new players in metabolic and cardiovascular disease. J Endocrinol 2016; 228:R57-71. [PMID: 26743452 DOI: 10.1530/joe-15-0201] [Citation(s) in RCA: 248] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/19/2015] [Indexed: 12/12/2022]
Abstract
The past decade has witnessed an exponential increase in the number of publications referring to extracellular vesicles (EVs). For many years considered to be extracellular debris, EVs are now seen as novel mediators of endocrine signalling via cell-to-cell communication. With the capability of transferring proteins and nucleic acids from one cell to another, they have become an attractive focus of research for different pathological settings and are now regarded as both mediators and biomarkers of disease including cardio-metabolic disease. They also offer therapeutic potential as signalling agents capable of targeting tissues or cells with specific peptides or miRNAs. In this review, we focus on the role that microvesicles (MVs) and exosomes, the two most studied classes of EV, have in diabetes, cardiovascular disease, endothelial dysfunction, coagulopathies, and polycystic ovary syndrome. We also provide an overview of current developments in MV/exosome isolation techniques from plasma and other fluids, comparing different available commercial and non-commercial methods. We describe different techniques for their optical/biochemical characterization and quantitation. We also review the signalling pathways that exosomes and MVs activate in target cells and provide some insight into their use as biomarkers or potential therapeutic agents. In summary, we give an updated focus on the role that these exciting novel nanoparticles offer for the endocrine community.
Collapse
Affiliation(s)
- Charlotte Lawson
- Department of Comparative Biomedical SciencesRoyal Veterinary College, Royal College Street, London NW1 0TU, UKThe Hatter Cardiovascular InstituteUniversity College London, London WC1E 6HX, UK
| | - Jose M Vicencio
- Department of Comparative Biomedical SciencesRoyal Veterinary College, Royal College Street, London NW1 0TU, UKThe Hatter Cardiovascular InstituteUniversity College London, London WC1E 6HX, UK
| | - Derek M Yellon
- Department of Comparative Biomedical SciencesRoyal Veterinary College, Royal College Street, London NW1 0TU, UKThe Hatter Cardiovascular InstituteUniversity College London, London WC1E 6HX, UK
| | - Sean M Davidson
- Department of Comparative Biomedical SciencesRoyal Veterinary College, Royal College Street, London NW1 0TU, UKThe Hatter Cardiovascular InstituteUniversity College London, London WC1E 6HX, UK
| |
Collapse
|
141
|
Erdbrügger U, Lannigan J. Analytical challenges of extracellular vesicle detection: A comparison of different techniques. Cytometry A 2015; 89:123-34. [PMID: 26651033 DOI: 10.1002/cyto.a.22795] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The interest in extracellular vesicles (EVs) has grown exponentially over the last decade. Evolving evidence is demonstrating that these EVs are playing an important role in health and disease. They are involved in intercellular communication and have been shown to transfer proteins, lipids, and nucleic acids. This review focuses on the most commonly used techniques for detection of EVs, to include microparticles, 100-1,000 nm in size, and exosomes, 50-100 nm in size. Conventional flow cytometry is the most prevalent technique, but nanoparticle tracking analysis (NTA), dynamic light scattering (DLS), and resistive pulse sensing have also been used to detect EVs. The accurate measurement of these vesicles is challenged by size heterogeneity, low refractive index, and the lack of dynamic measurement range for most of the available technologies. Sample handling during the preanalytical phase can also affect the accuracy of measurements. Currently, there is not one single method which allows phenotyping, sizing, and enumerating the whole range of EVs and, therefore, providing all the necessary information to truly understand the biology of these particles. A combination of methods is probably needed which might also include electron and atomic force microscopy and full RNA, lipid, and protein profiling.
Collapse
Affiliation(s)
- Uta Erdbrügger
- Department of Medicine, Division of Nephrology, University of Virginia Health System, Charlottesville, Virginia, 22908
| | - Joanne Lannigan
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, Virginia, 22908
| |
Collapse
|
142
|
Headland SE, Jones HR, Norling LV, Kim A, Souza PR, Corsiero E, Gil CD, Nerviani A, Dell'Accio F, Pitzalis C, Oliani SM, Jan LY, Perretti M. Neutrophil-derived microvesicles enter cartilage and protect the joint in inflammatory arthritis. Sci Transl Med 2015; 7:315ra190. [PMID: 26606969 PMCID: PMC6034622 DOI: 10.1126/scitranslmed.aac5608] [Citation(s) in RCA: 253] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Microvesicles (MVs) are emerging as a new mechanism of intercellular communication by transferring cellular lipid and protein components to target cells, yet their function in disease is only now being explored. We found that neutrophil-derived MVs were increased in concentration in synovial fluid from rheumatoid arthritis patients compared to paired plasma. Synovial MVs overexpressed the proresolving, anti-inflammatory protein annexin A1 (AnxA1). Mice deficient in TMEM16F, a lipid scramblase required for microvesiculation, exhibited exacerbated cartilage damage when subjected to inflammatory arthritis. To determine the function of MVs in inflammatory arthritis, toward the possibility of MV-based therapeutics, we examined the role of immune cell-derived MVs in rodent models and in human primary chondrocytes. In vitro, exogenous neutrophil-derived AnxA1(+) MVs activated anabolic gene expression in chondrocytes, leading to extracellular matrix accumulation and cartilage protection through the reduction in stress-adaptive homeostatic mediators interleukin-8 and prostaglandin E2. In vivo, intra-articular injection of AnxA1(+) MV lessened cartilage degradation caused by inflammatory arthritis. Arthritic mice receiving adoptive transfer of whole neutrophils displayed abundant MVs within cartilage matrix and revealed that MVs, but not neutrophils themselves, can penetrate cartilage. Mechanistic studies support a model whereby MV-associated AnxA1 interacts with its receptor FPR2 (formyl peptide receptor 2)/ALX, increasing transforming growth factor-β production by chondrocytes, ultimately leading to cartilage protection. We envisage that MVs, either directly or loaded with therapeutics, can be harnessed as a unique therapeutic strategy for protection in diseases associated with cartilage degeneration.
Collapse
Affiliation(s)
- Sarah E Headland
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Hefin R Jones
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Lucy V Norling
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Andrew Kim
- Department of Physiology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Patricia R Souza
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Elisa Corsiero
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Cristiane D Gil
- Department of Biology, Instituto de Biociências, Letras e Ciências Exatas, São Paulo State University (UNESP), São José do Rio Preto 15054-000, Brazil
| | - Alessandra Nerviani
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Francesco Dell'Accio
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK. Department of Rheumatology, Barts Health Trust, Bancroft Road, London E1 4DG, UK
| | - Costantino Pitzalis
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK. Department of Rheumatology, Barts Health Trust, Bancroft Road, London E1 4DG, UK
| | - Sonia M Oliani
- Department of Biology, Instituto de Biociências, Letras e Ciências Exatas, São Paulo State University (UNESP), São José do Rio Preto 15054-000, Brazil
| | - Lily Y Jan
- Department of Physiology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mauro Perretti
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
143
|
Immuno-analysis of microparticles: probing at the limits of detection. Sci Rep 2015; 5:16314. [PMID: 26553743 PMCID: PMC4639787 DOI: 10.1038/srep16314] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 10/07/2015] [Indexed: 12/20/2022] Open
Abstract
Microparticle (MP) research is clouded by debate regarding the accuracy and validity of flow cytometry (FCM) as an analytical methodology, as it is influenced by many variables including the pre-analytical conditions, instruments physical capabilities and detection parameters. This study utilises a simplistic in vitro system for generating MP, and through comparative analysis with immuno-electron microscopy (Immuno-EM) assesses the strengths and limitations of probe selection and high-sensitivity FCM. Of the markers examined, MP were most specifically labelled with phosphatidylserine ligands, annexin V and lactadherin, although only ~60% MP are PS positive. Whilst these two ligands detect comparable absolute MP numbers, they interact with the same population in distinct manners; annexin V binding is enhanced on TNF induced MP. CD105 and CD54 expression were, as expected, consistent and enhanced following TNF activation respectively. Their labelling however accounted for as few as 30-40% of MP. The greatest discrepancies between FCM and I-EM were observed in the population solely labelled for the surface antigen. These findings demonstrate that despite significant improvements in resolution, high-sensitivity FCM remains limited in detecting small-size MP expressing low antigen levels. This study highlights factors to consider when selecting endothelial MP probes, as well as interpreting and representing data.
Collapse
|
144
|
Bank IEM, Timmers L, Gijsberts CM, Zhang YN, Mosterd A, Wang JW, Chan MY, De Hoog V, Lim SK, Sze SK, Lam CSP, De Kleijn DPV. The diagnostic and prognostic potential of plasma extracellular vesicles for cardiovascular disease. Expert Rev Mol Diagn 2015; 15:1577-88. [DOI: 10.1586/14737159.2015.1109450] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
145
|
Stoner SA, Duggan E, Condello D, Guerrero A, Turk JR, Narayanan PK, Nolan JP. High sensitivity flow cytometry of membrane vesicles. Cytometry A 2015; 89:196-206. [PMID: 26484737 DOI: 10.1002/cyto.a.22787] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 07/31/2015] [Accepted: 09/23/2015] [Indexed: 12/17/2022]
Abstract
Extracellular vesicles (EVs) are attracting attention as vehicles for inter-cellular signaling that may have value as diagnostic or therapeutic targets. EVs are released by many cell types and by different mechanisms, resulting in phenotypic heterogeneity that makes them a challenge to study. Flow cytometry is a popular tool for characterizing heterogeneous mixtures of particles such as cell types within blood, but the small size of EVs makes them difficult to measure using conventional flow cytometry. To address this limitation, a high sensitivity flow cytometer was constructed and EV measurement approaches that allowed them to enumerate and estimate the size of individual EVs, as well as measure the presence of surface markers to identify phenotypic subsets of EVs. Several fluorescent membrane probes were evaluated and it was found that the voltage sensing dye di-8-ANEPPS could produce vesicle fluorescence in proportion to vesicle surface area, allowing for accurate measurements of EV number and size. Fluorescence-labeled annexin V and anti-CD61 antibody was used to measure the abundance of these surface markers on EVs in rat plasma. It was shown that treatment of platelet rich plasma with calcium ionophore resulted in an increase in the fraction of annexin V and CD61-positive EVs. Vesicle flow cytometry using fluorescence-based detection of EVs has the potential to realize the potential of cell-derived membrane vesicles as functional biomarkers for a variety of applications.
Collapse
Affiliation(s)
- Samuel A Stoner
- La Jolla Bioengineering Institute, La Jolla, California, 92037
| | - Erika Duggan
- La Jolla Bioengineering Institute, La Jolla, California, 92037.,Scintillon Institute, San Diego, California, 92121
| | - Danilo Condello
- La Jolla Bioengineering Institute, La Jolla, California, 92037.,Scintillon Institute, San Diego, California, 92121
| | - Abraham Guerrero
- Comparative Biology and Safety Sciences, Amgen Inc, Seattle, Washington
| | - James R Turk
- Comparative Biology and Safety Sciences, Amgen Inc, Seattle, Washington
| | - Padma K Narayanan
- Comparative Biology and Safety Sciences, Amgen Inc, Seattle, Washington
| | - John P Nolan
- La Jolla Bioengineering Institute, La Jolla, California, 92037.,Scintillon Institute, San Diego, California, 92121
| |
Collapse
|
146
|
Platelet microparticles in cryopreserved platelets: Potential mediators of haemostasis. Transfus Apher Sci 2015; 53:146-52. [DOI: 10.1016/j.transci.2015.10.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
147
|
|
148
|
Zhou BD, Guo G, Zheng LM, Zu LY, Gao W. Microparticles as novel biomarkers and therapeutic targets in coronary heart disease. Chin Med J (Engl) 2015; 128:267-72. [PMID: 25591573 PMCID: PMC4837849 DOI: 10.4103/0366-6999.149231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
| | | | | | - Ling-Yun Zu
- Department of Cardiology, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Third Hospital, Beijing 100191, China
| | | |
Collapse
|
149
|
Erdbrügger U, Le TH. Extracellular Vesicles in Renal Diseases: More than Novel Biomarkers? J Am Soc Nephrol 2015; 27:12-26. [PMID: 26251351 DOI: 10.1681/asn.2015010074] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles from the urine and circulation have gained significant interest as potential diagnostic biomarkers in renal diseases. Urinary extracellular vesicles contain proteins from all sections of the nephron, whereas most studied circulating extracellular vesicles are derived from platelets, immune cells, and the endothelium. In addition to their diagnostic role as markers of kidney and vascular damage, extracellular vesicles may have functional significance in renal health and disease by facilitating communication between cells and protecting against kidney injury and bacterial infection in the urinary tract. However, the current understanding of extracellular vesicles has derived mostly from studies with very small numbers of patients or in vitro data. Moreover, accurate assessment of these vesicles remains a challenge, in part because of a lack of consensus in the methodologies to measure extracellular vesicles and the inability of most techniques to capture the entire size range of these vesicles. However, newer techniques and standardized protocols to improve the detection of extracellular vesicles are in development. A clearer understanding of the composition and biology of extracellular vesicles will provide insights into their pathophysiologic, diagnostic, and therapeutic roles.
Collapse
Affiliation(s)
- Uta Erdbrügger
- Department of Medicine, Division of Nephrology, University of Virginia Health System, Charlottesville, Virginia
| | - Thu H Le
- Department of Medicine, Division of Nephrology, University of Virginia Health System, Charlottesville, Virginia
| |
Collapse
|
150
|
Extracellular vesicles as new pharmacological targets to treat atherosclerosis. Eur J Pharmacol 2015; 763:90-103. [PMID: 26142082 DOI: 10.1016/j.ejphar.2015.06.047] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 05/13/2015] [Accepted: 06/25/2015] [Indexed: 01/09/2023]
Abstract
Extracellular vesicles released by most cell types, include apoptotic bodies (ABs), microvesicles (MVs) and exosomes. They play a crucial role in physiology and pathology, contributing to "cell-to-cell" communication by modifying the phenotype and the function of target cells. Thus, extracellular vesicles participate in the key processes of atherosclerosis from endothelial dysfunction, vascular wall inflammation to vascular remodeling. The purpose of this review is to summarize recent findings on extracellular vesicle formation, structure, release and clearance. We focus on the deleterious and beneficial effects of extracellular vesicles in the development of atherosclerosis. The potential role of extracellular vesicles as biomarkers and pharmacological targets, their innate therapeutic capacity, or their use for novel drug delivery devices in atherosclerotic cardiovascular diseases will also be discussed.
Collapse
|