251
|
Yahata S, Hirose M, Ueno T, Nagumo H, Sakai-Kato K. Effect of Sample Concentration on Nanoparticle Tracking Analysis of Small Extracellular Vesicles and Liposomes Mimicking the Physicochemical Properties of Exosomes. Chem Pharm Bull (Tokyo) 2021; 69:1045-1053. [PMID: 34719585 DOI: 10.1248/cpb.c21-00452] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
For quantitative analysis, data should be obtained at a sample concentration that is within the range of linearity. We examined the effect of sample concentration on nanoparticle tracking analysis (NTA) of small extracellular vesicles (sEVs), including exosomes, by comparing NTA results of sEVs with those obtained for polystyrene nanoparticles (PSN) and liposomes, which mimic lipid composition and physicochemical properties of exosomes. Initially, NTA of PSN at different concentrations was performed and the particle sizes determined were validated by dynamic light scattering. The major peak maxima for PSN mixtures of different sizes at the higher particle numbers were similar, with some fluctuation of the minor peak maxima observed at the lower particle number, which was also observed for sEVs. Sample concentration is critical for obtaining reproducible data for liposomes and exosomes and increasing the sample concentration caused an increase in data variability because of particle interactions. The inter-day repeatability of particles sizes and concentration for sEVs were 7.47 and 4.51%, respectively. Analysis of the linearity range revealed that this was narrower for sEVs when compared with that of liposomes. Owing to the use of liposomes that mimic the lipid composition and physicochemical properties of exosomes and proteinase-treated sEVs, it was demonstrated that these different analytical results could be possibly caused by the protein corona of sEVs. Consideration of the sample concentration and linearity range is important for obtaining reproducible and reliable data of sEVs.
Collapse
|
252
|
Neyroud AS, Chiechio R, Yefimova M, Lo Faro MJ, Dejucq-Rainsford N, Jaillard S, Even-Hernandez P, Marchi V, Ravel C. Extra-cellular vesicles of the male genital tract: new actors in male fertility? Basic Clin Androl 2021; 31:25. [PMID: 34645388 PMCID: PMC8515699 DOI: 10.1186/s12610-021-00141-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular Vesicles (EVs) are membrane-limited particles containing proteins, lipids, metabolites and nucleic acids that are secreted by healthy and cancerous cells. These vesicles are very heterogeneous in size and content and mediate a variety of biological functions. Three subtypes of EV have been described in the male genital tract: microvesicles, myelinosomes and exosomes. Each type of EVs depends on the location of secretion such as the testis, prostate or epididymis. It has been shown that EVs can fuse together and deliver information to recipient cells, for example spermatozoa in the male genital tract. Cryo-electron microscopy remains the reference technique for determining EV morphology, but quantifying the absolute concentration of these EVs in biological fluids remains a challenge from a clinical point of view. The field of bio detection has considerably increased with the introduction of nanomaterials in biosensors and will provide a better understanding of the impact of these EVs. However, functional modifications of male gametes result from interactions with the components of the intraluminal fluid all along the genital tract and depend on the secretion and absorption of proteins and lipids from the local microenvironment. We cannot therefore exclude the possibility of epigenetic modulation of the information that will be transmitted to the embryo and therefore to the next generation via EVs.
Collapse
Affiliation(s)
- Anne-Sophie Neyroud
- CHU Rennes, Service de Biologie de la Reproduction-CECOS, 35000, Rennes, France
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Régina Chiechio
- Physics and Astronomy Department "E. Majorana", University of Catania, Via S. Sofia 64, 95123, Catania, Italy
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000, Rennes, France
| | - Marina Yefimova
- CHU Rennes, Service de Biologie de la Reproduction-CECOS, 35000, Rennes, France
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
- Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, St-Petersburg, 194223, Russia
| | - Maria Josè Lo Faro
- Physics and Astronomy Department "E. Majorana", University of Catania, Via S. Sofia 64, 95123, Catania, Italy
| | - Nathalie Dejucq-Rainsford
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Sylvie Jaillard
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Pascale Even-Hernandez
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000, Rennes, France
| | - Valérie Marchi
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000, Rennes, France
| | - Célia Ravel
- CHU Rennes, Service de Biologie de la Reproduction-CECOS, 35000, Rennes, France.
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France.
| |
Collapse
|
253
|
Midekessa G, Godakumara K, Dissanayake K, Hasan MM, Reshi QUA, Rinken T, Fazeli A. Characterization of Extracellular Vesicles Labelled with a Lipophilic Dye Using Fluorescence Nanoparticle Tracking Analysis. MEMBRANES 2021; 11:779. [PMID: 34677545 PMCID: PMC8539200 DOI: 10.3390/membranes11100779] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 12/05/2022]
Abstract
Research on extracellular vesicles (EVs) has intensified over the past decade, including fluorescent membrane labeling of EVs. An optimal fluorescent method requires the size of EVs to be preserved after labeling. Lipophilic fluorescent dyes, such as CellMask™ Green (CMG), have been widely used for this purpose. Here, we investigated conditions affecting the optimum CMG labeling of EVs derived from human choriocarcinoma cells (JAr) and different biological fluids using fluorescence NTA (fl-NTA). The effect of CMG labeling on the size, concentration and zeta potential (ZP) on JAr EVs purified with different methods were measured along with biological fluid-derived EVs. With the increase of CMG dye concentration, a significant decrease in the mean size of fluorescent nanoparticles (fl-NPs) was observed. The ZP of fl-NPs originating from JAr cells with the lowest and highest dye concentrations showed a significant shift towards more and less negative ZP values, respectively. Differences in the concentration of fl-NPs were observed for JAr EVs purified using size-exclusion chromatography (SEC) alone and SEC in combination with tangential flow filtration. The proportion of CMG labeling of NPs varied across different biological sources. CMG labeling may be a reliable technique for the detection of EVs using fl-NTA.
Collapse
Affiliation(s)
- Getnet Midekessa
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Kreutzwaldi 62, 51006 Tartu, Estonia; (G.M.); (K.G.); (K.D.); (M.M.H.); (Q.U.A.R.)
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 14b, 50411 Tartu, Estonia
| | - Kasun Godakumara
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Kreutzwaldi 62, 51006 Tartu, Estonia; (G.M.); (K.G.); (K.D.); (M.M.H.); (Q.U.A.R.)
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 14b, 50411 Tartu, Estonia
| | - Keerthie Dissanayake
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Kreutzwaldi 62, 51006 Tartu, Estonia; (G.M.); (K.G.); (K.D.); (M.M.H.); (Q.U.A.R.)
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 14b, 50411 Tartu, Estonia
- Department of Anatomy, Faculty of Medicine, University of Peradeniya, Peradeniya 20400, Sri Lanka
| | - Mohammad Mehedi Hasan
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Kreutzwaldi 62, 51006 Tartu, Estonia; (G.M.); (K.G.); (K.D.); (M.M.H.); (Q.U.A.R.)
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 14b, 50411 Tartu, Estonia
| | - Qurat Ul Ain Reshi
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Kreutzwaldi 62, 51006 Tartu, Estonia; (G.M.); (K.G.); (K.D.); (M.M.H.); (Q.U.A.R.)
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 14b, 50411 Tartu, Estonia
| | - Toonika Rinken
- Institute of Chemistry, University of Tartu, Ravila St. 14a, 50411 Tartu, Estonia;
| | - Alireza Fazeli
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Kreutzwaldi 62, 51006 Tartu, Estonia; (G.M.); (K.G.); (K.D.); (M.M.H.); (Q.U.A.R.)
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 14b, 50411 Tartu, Estonia
- Academic Unit of Reproductive and Developmental Medicine, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield S10 2SF, UK
| |
Collapse
|
254
|
Monnamorn L, Seree-Aphinan C, Molika P, Vichitkunakorn P, Pattanapanyasat K, Khwannimit B, Navakanitworakul R. The Concentration of Large Extracellular Vesicles Differentiates Early Septic Shock From Infection. Front Med (Lausanne) 2021; 8:724371. [PMID: 34604260 PMCID: PMC8481381 DOI: 10.3389/fmed.2021.724371] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/24/2021] [Indexed: 11/13/2022] Open
Abstract
Septic shock represents a subset of sepsis with severe physiological aberrations and a higher mortality rate than sepsis alone. Currently, the laboratory tools which can be used to identify the state of septic shock are limited. In pre-clinical studies, extracellular vesicles (EVs), especially large EVs (lEVs), have been demonstrated a role as functional inflammatory mediators of sepsis. However, its longitudinal trend during the disease course has not been explored. In this study, the quantities and subtypes of plasma-derived lEVs were longitudinally compared between patients with septic shock (n = 21) and non-sepsis infection (n = 9), who presented within 48 h of their symptom onset. Blood specimens were collected for seven consecutive days after hospital admission. lEVs quantification and subtyping were performed using an imaging flow cytometer. The experiments revealed a higher lEVs concentration in septic shock patients than infected patients at the onset of the disease. In septic shock patients, lEVs concentration decreased over time as opposed to infected patients whose lEVs concentration is relatively static throughout the study period. The major contributors of lEVs in both septic shock and infected patients were of non-leukocyte origins; platelets, erythrocytes, and endothelial cells released approximately 40, 25, and 15% of lEVs, respectively. Among lEVs of leukocyte origins, neutrophils produced the highest number of EVs. Nevertheless, the proportion of each subtype of lEVs among the given amount of lEVs produced was similar between septic shock and infected patients. These findings raise the possibility of employing lEVs enumeration as a septic shock identifying tool, although larger studies with a more diverse group of participants are warranted to extrapolate the findings to a general population.
Collapse
Affiliation(s)
- Latthawan Monnamorn
- Faculty of Medicine, Department of Biomedical Sciences and Biomedical Engineering, Prince of Songkla University, Songkhla, Thailand
| | - Chutima Seree-Aphinan
- Faculty of Medicine, Department of Internal Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Piyatida Molika
- Faculty of Medicine, Department of Biomedical Sciences and Biomedical Engineering, Prince of Songkla University, Songkhla, Thailand
| | - Polathep Vichitkunakorn
- Faculty of Medicine, Department of Family and Preventive Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Kovit Pattanapanyasat
- Faculty of Medicine Siriraj Hospital, Department of Research and Development, Mahidol University, Bangkok, Thailand
| | - Bodin Khwannimit
- Faculty of Medicine, Department of Internal Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Raphatphorn Navakanitworakul
- Faculty of Medicine, Department of Biomedical Sciences and Biomedical Engineering, Prince of Songkla University, Songkhla, Thailand
| |
Collapse
|
255
|
Zhu J, Zhang J, Ji X, Tan Z, Lubman DM. Column-based Technology for CD9-HPLC Immunoaffinity Isolation of Serum Extracellular Vesicles. J Proteome Res 2021; 20:4901-4911. [PMID: 34473505 DOI: 10.1021/acs.jproteome.1c00549] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Serum-derived extracellular vesicles (EVs) are a promising source of biomarkers; however, major challenges in EV separation and proteomic profiling remain for isolating EVs from a small amount, that is, on the microliter scale, of human serum while minimizing the contamination of blood proteins and lipoprotein particles coeluting in EV preparations. Herein we have developed a column-based CD9-antibody-immobilized high-performance liquid chromatography immunoaffinity chromatography(CD9-HPLC-IAC) technology for EV isolation from a microliter scale of serum for downstream proteomic analysis. The CD9-HPLC-IAC method achieved EV isolation from 40 μL of serum in 30 min with a yield of 8.0 × 109 EVs, where EVs were further processed with a postcolumn cleaning step using the 50 kDa molecular weight cut-off filter for the buffer exchange, concentration, and reduction of potentially coeluting serum proteins. In total, 482 proteins were identified in EVs by using liquid chromatography tandem mass spectrometry, including the common exosomal markers such as CD63, CD81, CD82, Alix, and TSG101. The statistical analysis of EV protein content showed that the top 10 serum proteins in EVs were significantly decreased by using the CD9-HPLC-IAC method compared with the use of ultracentrifugation (p = 0.001) and size exclusion chromatography (p = 0.009), and apolipoproteins were significantly reduced 4.8-fold compared with the SEC method (p < 0.001). The result demonstrates the potential of the CD9-HPLC-IAC method for the efficient isolation and proteomic characterization of EVs from a microscale volume of serum.
Collapse
Affiliation(s)
- Jianhui Zhu
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan 48109, United States
| | - Jie Zhang
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan 48109, United States
| | - Xiaohui Ji
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan 48109, United States.,Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Zhijing Tan
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan 48109, United States
| | - David M Lubman
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
256
|
Donzelli J, Proestler E, Riedel A, Nevermann S, Hertel B, Guenther A, Gattenlöhner S, Savai R, Larsson K, Saul MJ. Small extracellular vesicle-derived miR-574-5p regulates PGE2-biosynthesis via TLR7/8 in lung cancer. J Extracell Vesicles 2021; 10:e12143. [PMID: 34596365 PMCID: PMC8485338 DOI: 10.1002/jev2.12143] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/20/2021] [Accepted: 08/19/2021] [Indexed: 12/30/2022] Open
Abstract
Intercellular communication plays an essential role in lung cancer (LC). One of the major players in cell-cell-communication is small extracellular vesicles (sEV). SEV trigger various biological responses by transporting cellular cargo to target cells. One essential sEV component are microRNAs (miRs), whose transport has recently attracted increasing research interest. We report that prostaglandin E2 (PGE2 ), a key inflammatory lipid mediator, specifically induces the sorting of miR-574-5p in sEV of A549 and 2106T cells. We found that sEV-derived miR-574-5p activates Toll-like receptors (TLR) 7/8, thereby decreasing PGE2 -levels. In contrast, intracellular miR-574-5p induces PGE2 -biosynthesis. Consequently, the combination of intracellular and sEV-derived miR-574-5p controls PGE2 -levels via a feedback loop. This was only observed in adeno- but not in squamous cell carcinoma, indicating a cell-specific response to sEV-derived miRs, which might be due to unique tetraspanin compositions. Hence, we describe a novel function of miR-574-5p unique to adenocarcinoma. Intracellular miR-574-5p induces PGE2 and thus the secretion of sEV-derived miR-574-5p, which in turn decreases PGE2 -biosynthesis in recipient cells.
Collapse
Affiliation(s)
- Julia Donzelli
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| | - Eva Proestler
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| | - Anna Riedel
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| | - Sheila Nevermann
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| | - Brigitte Hertel
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| | - Andreas Guenther
- Department of Internal MedicineMember of the German Centre for Lung Research (DZL)Member of Cardio‐Pulmonary Institute (CPI)Justus Liebig UniversityGiessenGermany
| | | | - Rajkumar Savai
- Department of Internal MedicineMember of the German Centre for Lung Research (DZL)Member of Cardio‐Pulmonary Institute (CPI)Justus Liebig UniversityGiessenGermany
- Department of Lung Development and RemodellingMember of the DZLMember of CPIMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
- Lung Microenvironmental Niche in CancerogenesisInstitute for Lung Health (ILH)Justus Liebig UniversityGiessenGermany
| | - Karin Larsson
- Rheumatology UnitDepartment of MedicineKarolinska University HospitalStockholmSweden
| | - Meike J. Saul
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| |
Collapse
|
257
|
Kim HY, Song M, Gho YS, Kim H, Choi B. Extracellular vesicles derived from the periodontal pathogen Filifactor alocis induce systemic bone loss through Toll-like receptor 2. J Extracell Vesicles 2021; 10:e12157. [PMID: 34648247 PMCID: PMC8516034 DOI: 10.1002/jev2.12157] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/17/2021] [Accepted: 09/27/2021] [Indexed: 01/31/2023] Open
Abstract
Periodontitis is an inflammatory disease induced by local infection in tooth-supporting tissue. Periodontitis is associated with systemic bone diseases, but little is known about the mechanism of the causal effect of periodontitis on systemic bone resorption. Bacteria-derived extracellular vesicles (EVs) act as natural carriers of virulence factors that are responsible for systemic inflammation. In this study, we investigated the role of EVs derived from Filifactor alocis, a Gram-positive, anaerobic periodontal pathogen, in systemic bone loss and osteoclast differentiation. F. alocis EVs accumulated in the long bones of mice after intraperitoneal administration. These EVs induced proinflammatory cytokines, osteoclastogenesis, and bone resorption via Toll-like receptor 2 (TLR2). The phase separation of F. alocis EVs showed that amphiphilic molecules were responsible for the induced bone resorption and osteoclastogenesis. The osteoclastogenic effects of F. alocis EVs were reduced by lipoprotein lipase. Proteomic analysis of the amphiphilic molecules identified seven lipoproteins. Our results indicate that lipoprotein-like molecules in F. alocis EVs may contribute to systemic bone loss via TLR2.
Collapse
Affiliation(s)
- Hyun Young Kim
- Department of Oral Microbiology and ImmunologySchool of Dentistry, Seoul National UniversitySeoulRepublic of Korea
| | - Min‐Kyoung Song
- Department of Cell and Developmental BiologySchool of Dentistry, Seoul National UniversitySeoulRepublic of Korea
- Department of Internal MedicineSeoul National University HospitalSeoulRepublic of Korea
| | - Yong Song Gho
- Department of Life SciencesPohang University of Science and Technology (POSTECH)PohangRepublic of Korea
| | - Hong‐Hee Kim
- Department of Cell and Developmental BiologySchool of Dentistry, Seoul National UniversitySeoulRepublic of Korea
- Dental Research InstituteSeoul National UniversitySeoulRepublic of Korea
| | - Bong‐Kyu Choi
- Department of Oral Microbiology and ImmunologySchool of Dentistry, Seoul National UniversitySeoulRepublic of Korea
| |
Collapse
|
258
|
Fortunato D, Mladenović D, Criscuoli M, Loria F, Veiman KL, Zocco D, Koort K, Zarovni N. Opportunities and Pitfalls of Fluorescent Labeling Methodologies for Extracellular Vesicle Profiling on High-Resolution Single-Particle Platforms. Int J Mol Sci 2021; 22:10510. [PMID: 34638850 PMCID: PMC8508895 DOI: 10.3390/ijms221910510] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 12/26/2022] Open
Abstract
The relevance of extracellular vesicles (EVs) has grown exponentially, together with innovative basic research branches that feed medical and bioengineering applications. Such attraction has been fostered by the biological roles of EVs, as they carry biomolecules from any cell type to trigger systemic paracrine signaling or to dispose metabolism products. To fulfill their roles, EVs are transported through circulating biofluids, which can be exploited for the administration of therapeutic nanostructures or collected to intercept relevant EV-contained biomarkers. Despite their potential, EVs are ubiquitous and considerably heterogeneous. Therefore, it is fundamental to profile and identify subpopulations of interest. In this study, we optimized EV-labeling protocols on two different high-resolution single-particle platforms, the NanoFCM NanoAnalyzer (nFCM) and Particle Metrix ZetaView Fluorescence Nanoparticle Tracking Analyzer (F-NTA). In addition to the information obtained by particles' scattered light, purified and non-purified EVs from different cell sources were fluorescently stained with combinations of specific dyes and antibodies to facilitate their identification and characterization. Despite the validity and compatibility of EV-labeling strategies, they should be optimized for each platform. Since EVs can be easily confounded with similar-sized nanoparticles, it is imperative to control instrument settings and the specificity of staining protocols in order to conduct a rigorous and informative analysis.
Collapse
Affiliation(s)
| | - Danilo Mladenović
- HansaBioMed Life Sciences Ltd., 12618 Tallinn, Estonia; (D.M.); (F.L.); (K.-L.V.)
- School of Natural Sciences and Health, Tallinn University, 10120 Tallinn, Estonia;
| | | | - Francesca Loria
- HansaBioMed Life Sciences Ltd., 12618 Tallinn, Estonia; (D.M.); (F.L.); (K.-L.V.)
| | - Kadi-Liis Veiman
- HansaBioMed Life Sciences Ltd., 12618 Tallinn, Estonia; (D.M.); (F.L.); (K.-L.V.)
| | - Davide Zocco
- Exosomics SpA, 53100 Siena, Italy; (D.F.); (M.C.); (D.Z.)
- Cell and Gene Therapy Research and Development, Lonza Inc., Rockville, MD 20850, USA
| | - Kairi Koort
- School of Natural Sciences and Health, Tallinn University, 10120 Tallinn, Estonia;
| | - Natasa Zarovni
- Exosomics SpA, 53100 Siena, Italy; (D.F.); (M.C.); (D.Z.)
- HansaBioMed Life Sciences Ltd., 12618 Tallinn, Estonia; (D.M.); (F.L.); (K.-L.V.)
| |
Collapse
|
259
|
Hoffmann WH, Mulkerns NMC, Hall SR, Gersen H. Laser-induced convection shifts size distributions in nanoparticle tracking analysis. NANOSCALE ADVANCES 2021; 3:5694-5702. [PMID: 34604699 PMCID: PMC8478152 DOI: 10.1039/d1na00572c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/22/2021] [Indexed: 06/13/2023]
Abstract
This work discusses the effects of increasing laser power on the size data derived from NTA for particles of known size and scatterers in solutions of flufenamic acid in ethanol. We find that whilst a higher laser power reveals more particles as expected, their residence time changes due to laser-induced convection. This reduced residence time decreases the number of tracks available for individual particle size determination, shifting the size distribution to smaller values. This problem is overcome by using a shutter to inhibit the development of convection currents, increasing particle residence time and reducing the error on the size distribution. The detailed understanding of laser-induced convection permits more robust size characterisation of mesoscopic organic clusters, which play a key role in two-step nucleation theory.
Collapse
Affiliation(s)
- William H Hoffmann
- H. H. Wills Physics Laboratory, University of Bristol Tyndall Avenue Bristol BS8 1TL UK
- Bristol Centre for Functional Nanomaterials, University of Bristol Tyndall Avenue Bristol BS8 1TL UK
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
| | - Niall M C Mulkerns
- H. H. Wills Physics Laboratory, University of Bristol Tyndall Avenue Bristol BS8 1TL UK
- Bristol Centre for Functional Nanomaterials, University of Bristol Tyndall Avenue Bristol BS8 1TL UK
| | - Simon R Hall
- Bristol Centre for Functional Nanomaterials, University of Bristol Tyndall Avenue Bristol BS8 1TL UK
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
| | - Henkjan Gersen
- H. H. Wills Physics Laboratory, University of Bristol Tyndall Avenue Bristol BS8 1TL UK
- Bristol Centre for Functional Nanomaterials, University of Bristol Tyndall Avenue Bristol BS8 1TL UK
| |
Collapse
|
260
|
García-Flores M, Sánchez-López CM, Ramírez-Calvo M, Fernández-Serra A, Marcilla A, López-Guerrero JA. Isolation and characterization of urine microvesicles from prostate cancer patients: different approaches, different visions. BMC Urol 2021; 21:137. [PMID: 34579682 PMCID: PMC8477576 DOI: 10.1186/s12894-021-00902-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 09/20/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Because of their specific and biologically relevant cargo, urine extracellular vesicles (EVs) constitute a valuable source of potential non-invasive biomarkers that could support the clinical decision-making to improve the management of prostate cancer (PCa) patients. Different EV isolation methods differ in terms of complexity and yield, conditioning, as consequence, the analytical result. METHODS The aim of this study was to compare three different isolation methods for urine EVs: ultracentrifugation (UC), size exclusion chromatography (SEC), and a commercial kit (Exolute® Urine Kit). Urine samples were collected from 6 PCa patients and 4 healthy donors. After filtered through 0.22 µm filters, urine was divided in 3 equal volumes to perform EVs isolation with each of the three approaches. Isolated EVs were characterized by spectrophotometric protein quantification, nanoparticle tracking analysis, transmission electron microscopy, AlphaScreen Technology, and whole miRNA Transcriptome. RESULTS Our results showed that UC and SEC provided better results in terms of EVs yield and purity than Exolute®, non-significant differences were observed in terms of EV-size. Interestingly, luminescent AlphaScreen assay demonstrated a significant enrichment of CD9 and CD63 positive microvesicles in SEC and UC methods compared with Exolute®. This heterogeneity was also demonstrated in terms of miRNA content indicating that the best correlation was observed between UC and SEC. CONCLUSIONS Our study highlights the importance of standardizing the urine EV isolation methods to guaranty the analytical reproducibility necessary for their implementation in a clinical setting.
Collapse
Affiliation(s)
- María García-Flores
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, 46009, Valencia, Spain.,IVO-CIPF Joint Research Unit of Cancer, Príncipe Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - Christian M Sánchez-López
- Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, 46000, Burjassot, Valencia, Spain.,Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe, Universitat de Valencia, 46100, Valencia, Spain
| | - Marta Ramírez-Calvo
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, 46009, Valencia, Spain
| | - Antonio Fernández-Serra
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, 46009, Valencia, Spain
| | - Antonio Marcilla
- Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, 46000, Burjassot, Valencia, Spain. .,Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe, Universitat de Valencia, 46100, Valencia, Spain.
| | - José Antonio López-Guerrero
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, 46009, Valencia, Spain. .,IVO-CIPF Joint Research Unit of Cancer, Príncipe Felipe Research Center (CIPF), 46012, Valencia, Spain. .,Department of Pathology, School of Medicine, Catholic University of Valencia "San Vicente Mártir", 46001, Valencia, Spain.
| |
Collapse
|
261
|
Pham CV, Midge S, Barua H, Zhang Y, Ngoc-Gia Nguyen T, Barrero RA, Duan A, Yin W, Jiang G, Hou Y, Zhou S, Wang Y, Xie X, Tran PHL, Xiang D, Duan W. Bovine extracellular vesicles contaminate human extracellular vesicles produced in cell culture conditioned medium when 'exosome-depleted serum' is utilised. Arch Biochem Biophys 2021; 708:108963. [PMID: 34126088 DOI: 10.1016/j.abb.2021.108963] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 06/03/2021] [Accepted: 06/06/2021] [Indexed: 12/30/2022]
Abstract
Extracellular vesicles (EVs) are important intercellular communication messengers. Half of the published studies in the field are in vitro cell culture based in which bovine serum in various concentrations and forms is used to facilitate the production of extracellular vesicles. 'Exosome depleted serum' is the type of bovine serum most widely used in the production of human EVs. Herein, we demonstrate that, despite the initial caution raised in 2014 about the persistence of bovine EVs, 'exosome depleted serum' was still used in 46% of publications on human or rodent EVs between 2015 and 2019. Using nanoparticle tracking analysis combined with detergent lysis of vesicles as well as bovine CD9 ELISA, we show that there were approximately 5.33 x 107/mL of bovine EVs remaining in the 'exosome depleted serum'. Importantly, the 'exosome depleted serum' was relatively enriched in small EVs by approximately 2.7-fold relative to the large EVs compared to that in the original serum. Specifically, the percentage of small EVs in total vesicles had increased from the original 48% in the serum before ultracentrifugation to 92% in the 'exosome depleted serum'. Furthermore, the pervasive bovine EVs carried over by the 'exosome depleted serum', even when the lowest concentration (0.5%) was used in cell culture, resulted in a significant contamination of human EVs in cell culture conditioned medium. Our findings indicate that the use 'exosome depleted serum' in cell culture-based studies may introduce artefacts into research examining the function of human and rodent EVs, in particular those involving EV miRNA. Thus, we appeal to the researchers in the EV field to seriously reconsider the practice of using 'exosome depleted serum' in the production of human and other mammalian EVs in vitro.
Collapse
Affiliation(s)
- Cuong Viet Pham
- Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Victoria, 3216, Australia
| | - Snehal Midge
- Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Victoria, 3216, Australia
| | - Hridika Barua
- Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Victoria, 3216, Australia
| | - Yumei Zhang
- Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Victoria, 3216, Australia
| | - Tuong Ngoc-Gia Nguyen
- Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Victoria, 3216, Australia
| | - Roberto A Barrero
- eResearch, Division of Research and Innovation, Queensland University of Technology, 2 George Street, Brisbane City, QLD, 4000, Australia
| | - Andrew Duan
- School of Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University 27 Rainforest Walk, Clayton, VIC, 3800, Australia
| | - Wang Yin
- Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Victoria, 3216, Australia
| | - Guoqin Jiang
- Department of General Surgery, Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, 215004, PR China
| | - Yingchun Hou
- Laboratory of Tumor Molecular and Cellular Biology, College of Life Sciences, Shaanxi Normal University, 620 West Chang'an Avenue, Xi'an, Shaanxi, 710119, China
| | - Shufeng Zhou
- Department of Chemical Engineering & Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen, 361021, China
| | - Yiming Wang
- Shanghai OneTar Biomedicine, Shanghai, 201203, China
| | - Xiaoqing Xie
- Shanghai OneTar Biomedicine, Shanghai, 201203, China
| | - Phuong H L Tran
- Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Victoria, 3216, Australia.
| | - Dongxi Xiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, 200127, China; Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China; Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China.
| | - Wei Duan
- Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Victoria, 3216, Australia; Shanghai OneTar-Deakin Joint Laboratory of Personalized Precision Medicine, Shanghai, 201203, China.
| |
Collapse
|
262
|
Guo J, Wu C, Lin X, Zhou J, Zhang J, Zheng W, Wang T, Cui Y. Establishment of a simplified dichotomic size-exclusion chromatography for isolating extracellular vesicles toward clinical applications. J Extracell Vesicles 2021; 10:e12145. [PMID: 34514732 PMCID: PMC8435528 DOI: 10.1002/jev2.12145] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 08/03/2021] [Accepted: 08/31/2021] [Indexed: 02/06/2023] Open
Abstract
Size-exclusion chromatography (SEC) is a widely adopted method for the isolation of extracellular vesicles (EVs) from complex samples. SEC can efficiently remove high-abundant proteins, while often requires multiple fractionation operation using diversified column settings. In this study, we aim to establish a simplified SEC method to acquire high quality EVs. In comparison of all three cross-linked Sepharose resins with the sample types of FBS and human serum (HS), CL-6B and CL-4B showed superior performance in regular SEC to CL-2B in terms of significantly narrower EV and protein peaks, higher resolutions and EV purity. By increasing their bed volumes to 20 ml, the resolutions of CL-6B and CL-4B columns could be significantly improved, while the CL-6B column had the best performance with higher particle yields and tighter EV peaks. With the CL-6B 20 ml column, we further established a simplified dichotomic SEC method that only requires two bulk elutions to acquire EVs in the Eluate 1 and proteins in the Eluate 2. We further justified that such CL-6B columns were reusable for at least 10 consecutive times, and the dichotomic SEC was applicable to EV isolations from HS and FBS-free supernatants of fluorescently labelled and unlabelled SW620 cells. The proteomics analysis implicated that although the two methods had dissimilar abilities in removing different co-isolating contaminant proteins from EVs, the dichotomic SEC and ultracentrifugation could isolate EVs from human plasma with comparable purity. This dichotomic SEC has its intriguing potential to be used for EV preparation toward clinical testing and/or basic research.
Collapse
Affiliation(s)
- Jiahui Guo
- MOE Key Laboratory of Tumor Molecular BiologyKey Laboratory of Functional Protein Research of Guangdong Higher Education InstitutesInstitute of Life and Health EngineeringThe First Affiliated HospitalJinan UniversityGuangzhouGuangdongChina
| | - Caihong Wu
- MOE Key Laboratory of Tumor Molecular BiologyKey Laboratory of Functional Protein Research of Guangdong Higher Education InstitutesInstitute of Life and Health EngineeringThe First Affiliated HospitalJinan UniversityGuangzhouGuangdongChina
| | - Xinyi Lin
- MOE Key Laboratory of Tumor Molecular BiologyKey Laboratory of Functional Protein Research of Guangdong Higher Education InstitutesInstitute of Life and Health EngineeringThe First Affiliated HospitalJinan UniversityGuangzhouGuangdongChina
| | - Jian Zhou
- MOE Key Laboratory of Tumor Molecular BiologyKey Laboratory of Functional Protein Research of Guangdong Higher Education InstitutesInstitute of Life and Health EngineeringThe First Affiliated HospitalJinan UniversityGuangzhouGuangdongChina
| | - Jiayi Zhang
- MOE Key Laboratory of Tumor Molecular BiologyKey Laboratory of Functional Protein Research of Guangdong Higher Education InstitutesInstitute of Life and Health EngineeringThe First Affiliated HospitalJinan UniversityGuangzhouGuangdongChina
| | - Wenting Zheng
- MOE Key Laboratory of Tumor Molecular BiologyKey Laboratory of Functional Protein Research of Guangdong Higher Education InstitutesInstitute of Life and Health EngineeringThe First Affiliated HospitalJinan UniversityGuangzhouGuangdongChina
| | - Tong Wang
- MOE Key Laboratory of Tumor Molecular BiologyKey Laboratory of Functional Protein Research of Guangdong Higher Education InstitutesInstitute of Life and Health EngineeringThe First Affiliated HospitalJinan UniversityGuangzhouGuangdongChina
| | - Yizhi Cui
- MOE Key Laboratory of Tumor Molecular BiologyKey Laboratory of Functional Protein Research of Guangdong Higher Education InstitutesInstitute of Life and Health EngineeringThe First Affiliated HospitalJinan UniversityGuangzhouGuangdongChina
| |
Collapse
|
263
|
Extracellular Vesicle Separation Techniques Impact Results from Human Blood Samples: Considerations for Diagnostic Applications. Int J Mol Sci 2021; 22:ijms22179211. [PMID: 34502122 PMCID: PMC8431127 DOI: 10.3390/ijms22179211] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 01/14/2023] Open
Abstract
Extracellular vesicles (EVs) are reminiscent of their cell of origin and thus represent a valuable source of biomarkers. However, for EVs to be used as biomarkers in clinical practice, simple, comparable, and reproducible analytical methods must be applied. Although progress is being made in EV separation methods for human biofluids, the implementation of EV assays for clinical diagnosis and common guidelines are still lacking. We conducted a comprehensive analysis of established EV separation techniques from human serum and plasma, including ultracentrifugation and size exclusion chromatography (SEC), followed by concentration using (a) ultracentrifugation, (b) ultrafiltration, or (c) precipitation, and immunoaffinity isolation. We analyzed the size, number, protein, and miRNA content of the obtained EVs and assessed the functional delivery of EV cargo. Our results demonstrate that all methods led to an adequate yield of small EVs. While no significant difference in miRNA content was observed for the different separation methods, ultracentrifugation was best for subsequent flow cytometry analysis. Immunoaffinity isolation is not suitable for subsequent protein analyses. SEC + ultracentrifugation showed the best functional delivery of EV cargo. In summary, combining SEC with ultracentrifugation gives the highest yield of pure and functional EVs and allows reliable analysis of both protein and miRNA contents. We propose this combination as the preferred EV isolation method for biomarker studies from human serum or plasma.
Collapse
|
264
|
Zakeri A, Whitehead BJ, Stensballe A, de Korne C, Williams AR, Everts B, Nejsum P. Parasite worm antigens instruct macrophages to release immunoregulatory extracellular vesicles. J Extracell Vesicles 2021; 10:e12131. [PMID: 34429858 PMCID: PMC8365858 DOI: 10.1002/jev2.12131] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
Emerging evidence suggests that immune cells not only communicate with each other through cytokines, chemokines, and cell surface receptors, but also by releasing small membranous structures known as extracellular vesicles (EVs). EVs carry a variety of different molecules that can be taken up by recipient cells. Parasitic worms are well known for their immunomodulatory properties, but whether they can affect immune responses by altering EV-driven communication between host immune cells remains unclear. Here we provide evidence that stimulation of bone marrow-derived macrophages (BMDMs) with soluble products of Trichuris suis (TSPs), leads to the release of EVs with anti-inflammatory properties. Specifically, we found that EVs from TSP-pulsed BMDMs, but not those from unstimulated BMDMs can suppress TNFα and IL-6 release in LPS-stimulated BMDMs and BMDCs. However, no polarization toward M1 or M2 was observed in macrophages exposed to EVs. Moreover, EVs enhanced reactive oxygen species (ROS) production in the exposed BMDMs, which was associated with a deregulated redox homeostasis as revealed by pathway analysis of transcriptomic data. Proteomic analysis identified cytochrome p450 (CYP450) as a potential source of ROS in EVs from TSP-pulsed BMDMs. Finally, pharmacological inhibition of CYP450 activity could suppress ROS production in those BMDMs. In summary, we find that TSPs can modulate immune responses not only via direct interactions but also indirectly by eliciting the release of EVs from BMDMs that exert anti-inflammatory effects on recipient cells.
Collapse
Affiliation(s)
- Amin Zakeri
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | | | - Allan Stensballe
- Department of Medicine and Health TechnologyAalborg UniversityAalborgDenmark
| | - Clarize de Korne
- Department of ParasitologyLeiden University Medical CentreLeidenNetherlands
- Interventional Molecular Imaging laboratoryDepartment of RadiologyLeiden University Medical CentreLeidenNetherlands
| | - Andrew R. Williams
- Department of Veterinary and Animal SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenFrederiksbergDenmark
| | - Bart Everts
- Department of ParasitologyLeiden University Medical CentreLeidenNetherlands
| | - Peter Nejsum
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| |
Collapse
|
265
|
Rizzo J, Wong SSW, Gazi AD, Moyrand F, Chaze T, Commere P, Novault S, Matondo M, Péhau‐Arnaudet G, Reis FCG, Vos M, Alves LR, May RC, Nimrichter L, Rodrigues ML, Aimanianda V, Janbon G. Cryptococcus extracellular vesicles properties and their use as vaccine platforms. J Extracell Vesicles 2021; 10:e12129. [PMID: 34377375 PMCID: PMC8329992 DOI: 10.1002/jev2.12129] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 07/02/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
Whereas extracellular vesicle (EV) research has become commonplace in different biomedical fields, this field of research is still in its infancy in mycology. Here we provide a robust set of data regarding the structural and compositional aspects of EVs isolated from the fungal pathogenic species Cryptococcus neoformans, C. deneoformans and C. deuterogattii. Using cutting-edge methodological approaches including cryogenic electron microscopy and cryogenic electron tomography, proteomics, and flow cytometry, we revisited cryptococcal EV features and suggest a new EV structural model, in which the vesicular lipid bilayer is covered by mannoprotein-based fibrillar decoration, bearing the capsule polysaccharide as its outer layer. About 10% of the EV population is devoid of fibrillar decoration, adding another aspect to EV diversity. By analysing EV protein cargo from the three species, we characterized the typical Cryptococcus EV proteome. It contains several membrane-bound protein families, including some Tsh proteins bearing a SUR7/PalI motif. The presence of known protective antigens on the surface of Cryptococcus EVs, resembling the morphology of encapsulated virus structures, suggested their potential as a vaccine. Indeed, mice immunized with EVs obtained from an acapsular C. neoformans mutant strain rendered a strong antibody response in mice and significantly prolonged their survival upon C. neoformans infection.
Collapse
Affiliation(s)
- Juliana Rizzo
- Unité Biologie des ARN des Pathogènes FongiquesDépartement de Mycologie, Institut Pasteur, F‐75015ParisFrance
| | - Sarah Sze Wah Wong
- Unité Mycologie Moléculaire, CNRS UMR2000Département de Mycologie, Institut Pasteur, F‐75015ParisFrance
| | - Anastasia D. Gazi
- Ultrastructural Bio‐Imaging, UTechS UBI, CNRS UMR 3528Département de Biologie cellulaire et infection, Institut Pasteur, F‐75015ParisFrance
| | - Frédérique Moyrand
- Unité Biologie des ARN des Pathogènes FongiquesDépartement de Mycologie, Institut Pasteur, F‐75015ParisFrance
| | - Thibault Chaze
- Plateforme Protéomique, Unité de Spectrométrie de Masse pour la Biologie (MSBio), CNRS UMR 2000Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, F‐75015ParisFrance
| | - Pierre‐Henri Commere
- Cytometry and BiomarkersCentre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, F‐75015ParisFrance
| | - Sophie Novault
- Cytometry and BiomarkersCentre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, F‐75015ParisFrance
| | - Mariette Matondo
- Plateforme Protéomique, Unité de Spectrométrie de Masse pour la Biologie (MSBio), CNRS UMR 2000Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, F‐75015ParisFrance
| | - Gérard Péhau‐Arnaudet
- Ultrastructural Bio‐Imaging, UTechS UBI, CNRS UMR 3528Département de Biologie cellulaire et infection, Institut Pasteur, F‐75015ParisFrance
| | - Flavia C. G. Reis
- Instituto Carlos ChagasFundação Oswaldo Cruz (FIOCRUZ)CuritibaBrazil
- Centro de Desenvolvimento Tecnologico em Saude (CDTS‐Fiocruz)São PauloBrazil
| | - Matthijn Vos
- NanoImaging Core FacilityCentre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, F‐75015ParisFrance
| | | | - Robin C. May
- Institute of Microbiology and Infection and School of BiosciencesUniversity of BirminghamBirminghamB15 2TTUK
| | - Leonardo Nimrichter
- Instituto de Microbiologia Paulo de Góes (IMPG)Universidade Federal do Rio de JaneiroRio de JaneiroBrazil
| | - Marcio L. Rodrigues
- Instituto Carlos ChagasFundação Oswaldo Cruz (FIOCRUZ)CuritibaBrazil
- Instituto de Microbiologia Paulo de Góes (IMPG)Universidade Federal do Rio de JaneiroRio de JaneiroBrazil
| | - Vishukumar Aimanianda
- Unité Mycologie Moléculaire, CNRS UMR2000Département de Mycologie, Institut Pasteur, F‐75015ParisFrance
| | - Guilhem Janbon
- Unité Biologie des ARN des Pathogènes FongiquesDépartement de Mycologie, Institut Pasteur, F‐75015ParisFrance
| |
Collapse
|
266
|
Cho S, Yi J, Kwon Y, Kang H, Han C, Park J. Multifluorescence Single Extracellular Vesicle Analysis by Time-Sequential Illumination and Tracking. ACS NANO 2021; 15:11753-11761. [PMID: 34181396 DOI: 10.1021/acsnano.1c02556] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
We demonstrate a fluorescence-based nanoparticle tracking analysis (NTA) system for the characterization of both the size and membrane protein expression of individual extracellular vesicles (EVs). A sheet of lasers with four different wavelengths was sequentially shone onto extracellular vesicles according to a preprogrammed schedule, providing scattering images intercalated by three fluorescent images. The presence of extracellular vesicles was tracked frame by frame from scattering images. Fluorescence-labeled membrane proteins on EVs were detected by comparing scattering and fluorescent images. The tetraspanins (CD9, CD63, and CD81) of individual HEK293 EVs analyzed by both NTA and total internal reflection fluorescence microscopy showed that the proposed NTA system can contribute to the understanding of individual extracellular vesicles.
Collapse
Affiliation(s)
- Siwoo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro. Nam-Gu, Pohang, Gyeong-buk 37673, Republic of Korea
- EXOSOMEplus Inc., Suwon, Gyeonggi-do 16229, Republic of Korea
| | - Johan Yi
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro. Nam-Gu, Pohang, Gyeong-buk 37673, Republic of Korea
| | - Yongmin Kwon
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro. Nam-Gu, Pohang, Gyeong-buk 37673, Republic of Korea
| | - Hyejin Kang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, 77 Cheongam-Ro. Nam-Gu, Pohang, Gyeong-buk 37673, Republic of Korea
| | - Chungmin Han
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro. Nam-Gu, Pohang, Gyeong-buk 37673, Republic of Korea
| | - Jaesung Park
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro. Nam-Gu, Pohang, Gyeong-buk 37673, Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, 77 Cheongam-Ro. Nam-Gu, Pohang, Gyeong-buk 37673, Republic of Korea
| |
Collapse
|
267
|
Freitas de Lima F, da Silva BB, Oliveira JD, de Moura LD, Rodrigues da Silva GH, Fernandes PCL, Souza RIC, Dos Santos AC, de Paula E. Prolonged anesthesia and decreased toxicity of enantiomeric-excess bupivacaine loaded in ionic gradient liposomes. Int J Pharm 2021; 606:120944. [PMID: 34324985 DOI: 10.1016/j.ijpharm.2021.120944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 11/26/2022]
Abstract
Bupivacaine is the most employed local anesthetic in surgical procedures, worldwide. Its systemic toxicity has directed the synthesis of the less toxic, S(-) enantiomer. This work describes a formulation of ionic gradient liposomes (IGL) containing S75BVC, an enantiomeric excess mixture of 75% S(-) and 25% R(+) bupivacaine. IGL prepared with 250 mM (NH4)2SO4 in the inner aqueous core of phosphatidylcholine and cholesterol (3:2 mol%) vesicles plus 0.5% S75BVC showed average sizes of 312.5 ± 4.5 nm, low polydispersity (PDI < 0.18), negative zeta potentials (-14.2 ± 0.2 mV) and were stable for 360 days. The encapsulation efficiency achieved with IGLS75BVC (%EE = 38.6%) was higher than with IGL prepared with racemic bupivacaine (IGLRBVC, %EE = 28.3%). TEM images revealed spherical vesicles and µDSC analysis provided evidence on the interaction of the anesthetic with the lipid bilayer. Then, in vitro - release kinetics and cytotoxicity- and in vivo - toxic effects in Zebrafish and biochemical/histopathological analysis plus analgesia in Wistar rats - tests were performed. IGLS75BVC exhibited negligible toxicity against Schwann cells and Zebrafish larvae, and it did not affect biochemical markers or the morphology of rat tissues (heart, brain, cerebellum, sciatic nerve). The in vitro release of S75BVC from IGL was extended from 4 to 24 h, justifying the prolonged anesthetic effect measured in rats (~9 h). The advantages of IGLS75BVC formulation over IGLRBVC and plain bupivacaine formulations (prolonged anesthesia, preferential sensorial blockade, and no toxicity) confirm its potential for clinical use in surgical anesthesia.
Collapse
Affiliation(s)
- Fernando Freitas de Lima
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (Unicamp), Campinas, Brazil
| | - Bianca Brandão da Silva
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (Unicamp), Campinas, Brazil
| | - Juliana Damasceno Oliveira
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (Unicamp), Campinas, Brazil
| | - Ludmilla David de Moura
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (Unicamp), Campinas, Brazil
| | | | | | | | | | - Eneida de Paula
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (Unicamp), Campinas, Brazil.
| |
Collapse
|
268
|
Imanbekova M, Suarasan S, Rojalin T, Mizenko RR, Hilt S, Mathur M, Lepine P, Nicouleau M, Mohamed NV, Durcan TM, Carney RP, Voss JC, Wachsmann-Hogiu S. Identification of amyloid beta in small extracellular vesicles via Raman spectroscopy. NANOSCALE ADVANCES 2021; 3:4119-4132. [PMID: 34355118 PMCID: PMC8276787 DOI: 10.1039/d1na00330e] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/07/2021] [Indexed: 05/20/2023]
Abstract
One of the hallmarks of Alzheimer's disease (AD) pathogenesis is believed to be the production and deposition of amyloid-beta (Aβ) peptide into extracellular plaques. Existing research indicates that extracellular vesicles (EVs) can carry Aβ associated with AD. However, characterization of the EVs-associated Aβ and its conformational variants has yet to be realized. Raman spectroscopy is a label-free and non-destructive method that is able to assess the biochemical composition of EVs. This study reports for the first time the Raman spectroscopic fingerprint of the Aβ present in the molecular cargo of small extracellular vesicles (sEVs). Raman spectra were measured from sEVs isolated from Alzheimer's disease cell culture model, where secretion of Aβ is regulated by tetracycline promoter, and from midbrain organoids. The averaged spectra of each sEV group showed considerable variation as a reflection of the biochemical content of sEVs. Spectral analysis identified more intense Raman peaks at 1650 cm-1 and 2930 cm-1 attributable to the Aβ peptide incorporated in sEVs produced by the Alzheimer's cell culture model. Subsequent analysis of the spectra by principal component analysis differentiated the sEVs of the Alzheimer's disease cell culture model from the control groups of sEVs. Moreover, the results indicate that Aβ associated with secreted sEVs has a α-helical secondary structure and the size of a monomer or small oligomer. Furthermore, by analyzing the lipid content of sEVs we identified altered fatty acid chain lengths in sEVs that carry Aβ that may affect the fluidity of the EV membrane. Overall, our findings provide evidence supporting the use of Raman spectroscopy for the identification and characterization of sEVs associated with potential biomarkers of neurological disorders such as toxic proteins.
Collapse
Affiliation(s)
| | - Sorina Suarasan
- Department of Bioengineering, McGill University Montreal QC H3A 0E9 Canada
| | - Tatu Rojalin
- Department of Biomedical Engineering, University of California Davis CA 95616 USA
| | - Rachel R Mizenko
- Department of Biomedical Engineering, University of California Davis CA 95616 USA
| | - Silvia Hilt
- Department of Biochemistry & Molecular Medicine, University of California Davis CA 95616 USA
| | - Meghna Mathur
- The Early Drug Discovery Unit (EDDU), Montreal Neurological Institute and Hospital, McGill University Montreal QC H3A 2B4 Canada
| | - Paula Lepine
- The Early Drug Discovery Unit (EDDU), Montreal Neurological Institute and Hospital, McGill University Montreal QC H3A 2B4 Canada
| | - Michael Nicouleau
- The Early Drug Discovery Unit (EDDU), Montreal Neurological Institute and Hospital, McGill University Montreal QC H3A 2B4 Canada
| | - Nguyen-Vi Mohamed
- The Early Drug Discovery Unit (EDDU), Montreal Neurological Institute and Hospital, McGill University Montreal QC H3A 2B4 Canada
| | - Thomas M Durcan
- The Early Drug Discovery Unit (EDDU), Montreal Neurological Institute and Hospital, McGill University Montreal QC H3A 2B4 Canada
| | - Randy P Carney
- Department of Biomedical Engineering, University of California Davis CA 95616 USA
| | - John C Voss
- Department of Biochemistry & Molecular Medicine, University of California Davis CA 95616 USA
| | | |
Collapse
|
269
|
Veerman RE, Teeuwen L, Czarnewski P, Güclüler Akpinar G, Sandberg A, Cao X, Pernemalm M, Orre LM, Gabrielsson S, Eldh M. Molecular evaluation of five different isolation methods for extracellular vesicles reveals different clinical applicability and subcellular origin. J Extracell Vesicles 2021; 10:e12128. [PMID: 34322205 PMCID: PMC8298890 DOI: 10.1002/jev2.12128] [Citation(s) in RCA: 181] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/21/2021] [Accepted: 07/13/2021] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are increasingly tested as therapeutic vehicles and biomarkers, but still EV subtypes are not fully characterised. To isolate EVs with few co-isolated entities, a combination of methods is needed. However, this is time-consuming and requires large sample volumes, often not feasible in most clinical studies or in studies where small sample volumes are available. Therefore, we compared EVs rendered by five commonly used methods based on different principles from conditioned cell medium and 250 μl or 3 ml plasma, that is, precipitation (ExoQuick ULTRA), membrane affinity (exoEasy Maxi Kit), size-exclusion chromatography (qEVoriginal), iodixanol gradient (OptiPrep), and phosphatidylserine affinity (MagCapture). EVs were characterised by electron microscopy, Nanoparticle Tracking Analysis, Bioanalyzer, flow cytometry, and LC-MS/MS. The different methods yielded samples of different morphology, particle size, and proteomic profile. For the conditioned medium, Izon 35 isolated the highest number of EV proteins followed by exoEasy, which also isolated fewer non-EV proteins. For the plasma samples, exoEasy isolated a high number of EV proteins and few non-EV proteins, while Izon 70 isolated the most EV proteins. We conclude that no method is perfect for all studies, rather, different methods are suited depending on sample type and interest in EV subtype, in addition to sample volume and budget.
Collapse
Affiliation(s)
- Rosanne E. Veerman
- Department of Clinical Immunology and Transfusion Medicine and Division of Immunology and Allergy, Department of Medicine SolnaKarolinska University Hospital and Karolinska InstitutetStockholmSweden
| | - Loes Teeuwen
- Department of Clinical Immunology and Transfusion Medicine and Division of Immunology and Allergy, Department of Medicine SolnaKarolinska University Hospital and Karolinska InstitutetStockholmSweden
| | - Paulo Czarnewski
- Science for Life LaboratoryDepartment of Biochemistry and BiophysicsNational Bioinformatics Infrastructure SwedenStockholm UniversitySolnaSweden
| | - Gözde Güclüler Akpinar
- Department of Clinical Immunology and Transfusion Medicine and Division of Immunology and Allergy, Department of Medicine SolnaKarolinska University Hospital and Karolinska InstitutetStockholmSweden
| | - AnnSofi Sandberg
- Department of Oncology and PathologyKarolinska InstitutetScience for Life LaboratorySolnaSweden
| | - Xiaofang Cao
- Department of Oncology and PathologyKarolinska InstitutetScience for Life LaboratorySolnaSweden
| | - Maria Pernemalm
- Department of Oncology and PathologyKarolinska InstitutetScience for Life LaboratorySolnaSweden
| | - Lukas M. Orre
- Department of Oncology and PathologyKarolinska InstitutetScience for Life LaboratorySolnaSweden
| | - Susanne Gabrielsson
- Department of Clinical Immunology and Transfusion Medicine and Division of Immunology and Allergy, Department of Medicine SolnaKarolinska University Hospital and Karolinska InstitutetStockholmSweden
| | - Maria Eldh
- Department of Clinical Immunology and Transfusion Medicine and Division of Immunology and Allergy, Department of Medicine SolnaKarolinska University Hospital and Karolinska InstitutetStockholmSweden
| |
Collapse
|
270
|
Phillips W, Willms E, Hill AF. Understanding extracellular vesicle and nanoparticle heterogeneity: Novel methods and considerations. Proteomics 2021; 21:e2000118. [PMID: 33857352 PMCID: PMC8365743 DOI: 10.1002/pmic.202000118] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/22/2021] [Accepted: 04/12/2021] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EVs) are a heterogeneous population of membrane-enclosed nanoparticles released by cells. They play a role in intercellular communication and are involved in numerous physiological and pathological processes. Cells release subpopulations of EVs with distinct composition and inherent biological function which overlap in size. Current size-based isolation methods are, therefore, not optimal to discriminate between functional EV subpopulations. In addition, EVs overlap in size with several other biological nanoparticles, such as lipoproteins and viruses. Proteomic analysis has allowed for more detailed study of EV composition, and EV isolation approaches based on this could provide a promising alternative for purification based on size. Elucidating EV heterogeneity and the characteristics and role of EV subpopulations will advance our understanding of EV biology and the role of EVs in health and disease. Here, we discuss current knowledge of EV composition, EV heterogeneity and advances in affinity based EV isolation tools.
Collapse
Affiliation(s)
- William Phillips
- Department of Biochemistry and GeneticsLa Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVictoriaAustralia
| | - Eduard Willms
- Department of Biochemistry and GeneticsLa Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVictoriaAustralia
| | - Andrew F. Hill
- Department of Biochemistry and GeneticsLa Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVictoriaAustralia
| |
Collapse
|
271
|
Buschmann D, Mussack V, Byrd JB. Separation, characterization, and standardization of extracellular vesicles for drug delivery applications. Adv Drug Deliv Rev 2021; 174:348-368. [PMID: 33964356 PMCID: PMC8217305 DOI: 10.1016/j.addr.2021.04.027] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/25/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles (EVs) are membranous nanovesicles secreted from living cells, shuttling macromolecules in intercellular communication and potentially possessing intrinsic therapeutic activity. Due to their stability, low immunogenicity, and inherent interaction with recipient cells, EVs also hold great promise as drug delivery vehicles. Indeed, they have been used to deliver nucleic acids, proteins, and small molecules in preclinical investigations. Furthermore, EV-based drugs have entered early clinical trials for cancer or neurodegenerative diseases. Despite their appeal as delivery vectors, however, EV-based drug delivery progress has been hampered by heterogeneity of sample types and methods as well as a persistent lack of standardization, validation, and comprehensive reporting. This review highlights specific requirements for EVs in drug delivery and describes the most pertinent approaches for separation and characterization. Despite residual uncertainties related to pharmacodynamics, pharmacokinetics, and potential off-target effects, clinical-grade, high-potency EV drugs might be achievable through GMP-compliant workflows in a highly standardized environment.
Collapse
Affiliation(s)
- Dominik Buschmann
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Veronika Mussack
- Department of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - James Brian Byrd
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
272
|
Gualerzi A, Picciolini S, Carlomagno C, Rodà F, Bedoni M. Biophotonics for diagnostic detection of extracellular vesicles. Adv Drug Deliv Rev 2021; 174:229-249. [PMID: 33887403 DOI: 10.1016/j.addr.2021.04.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/26/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023]
Abstract
Extracellular Vesicles (EVs) are versatile carriers for biomarkers involved in the pathogenesis of multiple human disorders. Despite the increasing scientific and commercial interest in EV application in diagnostics, traditional biomolecular techniques usually require consistent sample amount, rely on operator-dependent and time- consuming procedures and cannot cope with the nano-size range of EVs, limiting both sensitivity and reproducibility of results. The application of biophotonics, i.e. light-based methods, for the diagnostic detection of EVs has brought to the development of innovative platforms with excellent sensitivity. In this review, we propose an overview of the most promising and emerging technologies used in the field of EV-related biomarker discovery. When tested on clinical samples, the reported biophotonic approaches in most cases have managed to discriminate between nanovesicles and contaminants, achieved much higher resolution compared to traditional procedures, and reached moderate to excellent diagnostic accuracy, thus demonstrating great potentialities for their clinical translation.
Collapse
|
273
|
Tsai SCS, Yang KD, Chang KH, Lin FCF, Chou RH, Li MC, Cheng CC, Kao CY, Chen CP, Lin HC, Hsu YC. Umbilical Cord Mesenchymal Stromal Cell-Derived Exosomes Rescue the Loss of Outer Hair Cells and Repair Cochlear Damage in Cisplatin-Injected Mice. Int J Mol Sci 2021; 22:ijms22136664. [PMID: 34206364 PMCID: PMC8267798 DOI: 10.3390/ijms22136664] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/10/2021] [Accepted: 06/12/2021] [Indexed: 12/21/2022] Open
Abstract
Umbilical cord-derived mesenchymal stromal cells (UCMSCs) have potential applications in regenerative medicine. UCMSCs have been demonstrated to repair tissue damage in many inflammatory and degenerative diseases. We have previously shown that UCMSC exosomes reduce nerve injury-induced pain in rats. In this study, we characterized UCMSC exosomes using RNA sequencing and proteomic analyses and investigated their protective effects on cisplatin-induced hearing loss in mice. Two independent experiments were designed to investigate the protective effects on cisplatin-induced hearing loss in mice: (i) chronic intraperitoneal cisplatin administration (4 mg/kg) once per day for 5 consecutive days and intraperitoneal UCMSC exosome (1.2 μg/μL) injection at the same time point; and (ii) UCMSC exosome (1.2 μg/μL) injection through a round window niche 3 days after chronic cisplatin administration. Our data suggest that UCMSC exosomes exert protective effects in vivo. The post-traumatic administration of UCMSC exosomes significantly improved hearing loss and rescued the loss of cochlear hair cells in mice receiving chronic cisplatin injection. Neuropathological gene panel analyses further revealed the UCMSC exosomes treatment led to beneficial changes in the expression levels of many genes in the cochlear tissues of cisplatin-injected mice. In conclusion, UCMSC exosomes exerted protective effects in treating ototoxicity-induced hearing loss by promoting tissue remodeling and repair.
Collapse
Affiliation(s)
- Stella Chin-Shaw Tsai
- Department of Otolaryngology, Tungs’ Taichung Metroharbor Hospital, Taichung 435, Taiwan;
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, 145, Guoguang Rd., South Dist., Taichung City 402, Taiwan
| | - Kuender D. Yang
- Department of Medical Research, Mackay Memorial Hospital, Taipei 104, Taiwan; (K.D.Y.); (C.-P.C.)
- Department of Otolaryngology, Mackay Memorial Hospital, New Taipei City 251, Taiwan
| | - Kuang-Hsi Chang
- Department of Medical Research, Tungs’ Taichung Metroharbor Hospital, Taichung 435, Taiwan;
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406, Taiwan;
- General Education Center, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 356, Taiwan
| | - Frank Cheau-Feng Lin
- Department of Thoracic Surgery, Chung Shan Medical University Hospital, Taichung 402, Taiwan;
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Ruey-Hwang Chou
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406, Taiwan;
- Center for Molecular Medicine, China Medical University Hospital, Taichung 406, Taiwan
- Department of Medical Laboratory and Biotechnology, Asia University, Taichung 413, Taiwan
| | - Min-Chih Li
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City 252, Taiwan;
| | - Ching-Chang Cheng
- Laboratory Animal Service Center, Office of Research and Development, China Medical University, Taichung 406, Taiwan;
| | - Chien-Yu Kao
- Medical and Pharmaceutical Industry Technology and Development Center, New Taipei City 248, Taiwan;
| | - Chie-Pein Chen
- Department of Medical Research, Mackay Memorial Hospital, Taipei 104, Taiwan; (K.D.Y.); (C.-P.C.)
| | - Hung-Ching Lin
- Department of Audiology and Speech-Language Pathology, Mackay Medical College, New Taipei City 252, Taiwan;
| | - Yi-Chao Hsu
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City 252, Taiwan;
- Department of Audiology and Speech-Language Pathology, Mackay Medical College, New Taipei City 252, Taiwan;
- Correspondence: ; Tel.: +886-2-26360303 (ext. 1721)
| |
Collapse
|
274
|
Levy D, Do MA, Zhang J, Brown A, Lu B. Orchestrating Extracellular Vesicle With Dual Reporters for Imaging and Capturing in Mammalian Cell Culture. Front Mol Biosci 2021; 8:680580. [PMID: 34222335 PMCID: PMC8249585 DOI: 10.3389/fmolb.2021.680580] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 05/31/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Recent technological advancements have enabled live-cell imaging of intracellular organelles to monitor their biogenesis in mammalian cells. However, applying this method to gain insight into extracellular organelles, such as extracellular vesicles (EVs), presents unique challenges that require special considerations in design and engineering. Results: We have developed a dual-reporter system that combines genetic fusion, fluorescence microcopy and magnetic beads capture of EVs to study the biogenesis of EVs in mammalian cell cultures. First, we genetically produced a series of reporters by fusing a green fluorescent protein (GFP) and an affinity peptide (6xHis), with either the endogenous transmembrane protein, CD63, or EVs targeting vesicular stomatitis viral glycoprotein (VSVG). Transfection of these reporters into human 293T cells resulted in expression and integration of these reporters into pre-exosome compartments, which were subsequently released into the culture medium. Confocal imaging and nano-particle tracking analysis demonstrated that EVs were appropriately labeled and exhibited a single dominant peak in the 80–110 nm size range, indicating that isolated EVs were comprised of micro-vesicles and/or exosome subpopulations. Incubation of isolated EVs with nickel-coated magnetic beads resulted in successful capture of GFP-positive EVs. Finally, addition of EVs into culture medium was able to reveal the cellular uptake of GFP-labeled EVs by recipient cells. Taken together, our dual-reporter system provides a powerful method for both monitoring and capturing of EVs in mammalian cell culture systems. Conclusion: A dual-reporter system provides a robust tool to study the life cycle of EVs in mammalian cells from biogenesis and excretion to cellular uptake.
Collapse
Affiliation(s)
- Daniel Levy
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA, United States
| | - Mai Anh Do
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA, United States
| | - Jiayi Zhang
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA, United States
| | - Annie Brown
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA, United States
| | - Biao Lu
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA, United States
| |
Collapse
|
275
|
Coughlan C, Bruce KD, Burgy O, Boyd TD, Michel CR, Garcia-Perez JE, Adame V, Anton P, Bettcher BM, Chial HJ, Königshoff M, Hsieh EWY, Graner M, Potter H. Exosome Isolation by Ultracentrifugation and Precipitation and Techniques for Downstream Analyses. ACTA ACUST UNITED AC 2021; 88:e110. [PMID: 32633898 DOI: 10.1002/cpcb.110] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Exosomes are 50- to 150-nm-diameter extracellular vesicles secreted by all mammalian cells except mature red blood cells and contribute to diverse physiological and pathological functions within the body. Many methods have been used to isolate and analyze exosomes, resulting in inconsistencies across experiments and raising questions about how to compare results obtained using different approaches. Questions have also been raised regarding the purity of the various preparations with regard to the sizes and types of vesicles and to the presence of lipoproteins. Thus, investigators often find it challenging to identify the optimal exosome isolation protocol for their experimental needs. Our laboratories have compared ultracentrifugation and commercial precipitation- and column-based exosome isolation kits for exosome preparation. Here, we present protocols for exosome isolation using two of the most commonly used methods, ultracentrifugation and precipitation, followed by downstream analyses. We use NanoSight nanoparticle tracking analysis and flow cytometry (Cytek® ) to determine exosome concentrations and sizes. Imaging flow cytometry can be utilized to both size exosomes and immunophenotype surface markers on exosomes (ImageStream® ). High-performance liquid chromatography followed by nano-flow liquid chromatography-mass spectrometry (LCMS) of the exosome fractions can be used to determine the presence of lipoproteins, with LCMS able to provide a proteomic profile of the exosome preparations. We found that the precipitation method was six times faster and resulted in a ∼2.5-fold higher concentration of exosomes per milliliter compared to ultracentrifugation. Both methods yielded extracellular vesicles in the size range of exosomes, and both preparations included apoproteins. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Pre-analytic fluid collection and processing Basic Protocol 2: Exosome isolation by ultracentrifugation Alternate Protocol 1: Exosome isolation by precipitation Basic Protocol 3: Analysis of exosomes by NanoSight nanoparticle tracking analysis Alternate Protocol 2: Analysis of exosomes by flow cytometry and imaging flow cytometry Basic Protocol 4: Downstream analysis of exosomes using high-performance liquid chromatography Basic Protocol 5: Downstream analysis of the exosome proteome using nano-flow liquid chromatography-mass spectrometry.
Collapse
Affiliation(s)
- Christina Coughlan
- University of Colorado Alzheimer's and Cognition Center, Department of Neurology, Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Kimberley D Bruce
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Olivier Burgy
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus. INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, Dijon, France
| | - Timothy D Boyd
- University of Colorado Alzheimer's and Cognition Center, Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Cole R Michel
- School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Josselyn E Garcia-Perez
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Vanesa Adame
- University of Colorado Alzheimer's and Cognition Center, Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Paige Anton
- Department of Pharmaceutical Sciences, University of Colorado Alzheimer's and Cognition Center, Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Brianne M Bettcher
- University of Colorado Alzheimer's and Cognition Center, Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Heidi J Chial
- University of Colorado Alzheimer's and Cognition Center, Department of Neurology, Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Melanie Königshoff
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Elena W Y Hsieh
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado
| | - Michael Graner
- Department of Pediatrics, Section of Allergy and Immunology, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, Colorado
| | - Huntington Potter
- University of Colorado Alzheimer's and Cognition Center, Department of Neurology, Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
276
|
Bordanaba-Florit G, Royo F, Kruglik SG, Falcón-Pérez JM. Using single-vesicle technologies to unravel the heterogeneity of extracellular vesicles. Nat Protoc 2021; 16:3163-3185. [PMID: 34135505 DOI: 10.1038/s41596-021-00551-z] [Citation(s) in RCA: 166] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 03/31/2021] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles (EVs) are heterogeneous lipid containers with a complex molecular cargo comprising several populations with unique roles in biological processes. These vesicles are closely associated with specific physiological features, which makes them invaluable in the detection and monitoring of various diseases. EVs play a key role in pathophysiological processes by actively triggering genetic or metabolic responses. However, the heterogeneity of their structure and composition hinders their application in medical diagnosis and therapies. This diversity makes it difficult to establish their exact physiological roles, and the functions and composition of different EV (sub)populations. Ensemble averaging approaches currently employed for EV characterization, such as western blotting or 'omics' technologies, tend to obscure rather than reveal these heterogeneities. Recent developments in single-vesicle analysis have made it possible to overcome these limitations and have facilitated the development of practical clinical applications. In this review, we discuss the benefits and challenges inherent to the current methods for the analysis of single vesicles and review the contribution of these approaches to the understanding of EV biology. We describe the contributions of these recent technological advances to the characterization and phenotyping of EVs, examination of the role of EVs in cell-to-cell communication pathways and the identification and validation of EVs as disease biomarkers. Finally, we discuss the potential of innovative single-vesicle imaging and analysis methodologies using microfluidic devices, which promise to deliver rapid and effective basic and practical applications for minimally invasive prognosis systems.
Collapse
Affiliation(s)
- Guillermo Bordanaba-Florit
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain.
| | - Félix Royo
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Madrid, Spain
| | - Sergei G Kruglik
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Laboratoire Jean Perrin, Paris, France
| | - Juan M Falcón-Pérez
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Madrid, Spain. .,Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
277
|
Brown HL, Clayton A, Stephens P. The role of bacterial extracellular vesicles in chronic wound infections: Current knowledge and future challenges. Wound Repair Regen 2021; 29:864-880. [PMID: 34132443 DOI: 10.1111/wrr.12949] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/14/2021] [Accepted: 05/26/2021] [Indexed: 12/16/2022]
Abstract
Chronic wounds are a significant global problem with an increasing economic and patient welfare impact. How wounds move from an acute to chronic, non-healing, state is not well understood although it is likely that it is driven by a poorly regulated local inflammatory state. Opportunistic pathogens such as Staphylococcus aureus and Pseudomonas aeruginosa are well known to stimulate a pro-inflammatory response and so their presence may further drive chronicity. Studies have demonstrated that host cell extracellular vesicles (hEVs), in particular exosomes, have multiple roles in both increasing and decreasing chronicity within wounds; however, the role of bacterial extracellular vesicles (bEVs) is still poorly understood. The aim of this review is to evaluate bEV biogenesis and function within chronic wound relevant bacterial species to determine what, if any, role bEVs may have in driving wound chronicity. We determine that bEVs drive chronicity by both increasing persistence of key pathogens such as Staphylococcus aureus and Pseudomonas aeruginosa and stimulating a pro-inflammatory response by the host. Data also suggest that both bEVs and hEVs show therapeutic promise, providing vaccine candidates, decoy targets for bacterial toxins or modulating the bacterial species within chronic wound biofilms. Caution should, however, be used when interpreting findings to date as the bEV field is still in its infancy and as such lacks consistency in bEV isolation and characterization. It is of primary importance that this is addressed, allowing meaningful conclusions to be drawn and increasing reproducibility within the field.
Collapse
Affiliation(s)
- Helen L Brown
- School of Dentistry, Cardiff University, Cardiff, UK
| | - Aled Clayton
- Division of Cancer & Genetics, School of Medicine, Cardiff, UK
| | - Phil Stephens
- School of Dentistry, Cardiff University, Cardiff, UK
| |
Collapse
|
278
|
López de Las Hazas MC, Gil-Zamorano J, Cofán M, Mantilla-Escalante DC, Garcia-Ruiz A, Del Pozo-Acebo L, Pastor O, Yañez-Mo M, Mazzeo C, Serra-Mir M, Doménech M, Valls-Pedret C, Rajaram S, Sabaté J, Ros E, Sala-Vila A, Dávalos A. One-year dietary supplementation with walnuts modifies exosomal miRNA in elderly subjects. Eur J Nutr 2021; 60:1999-2011. [PMID: 32979076 DOI: 10.1007/s00394-020-02390-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE Epidemiological studies and clinical trials support the association of nut consumption with a lower risk of prevalent non-communicable diseases, particularly cardiovascular disease. However, the molecular mechanisms underlying nut benefits remain to be fully described. MicroRNAs (miRNAs) are post-transcriptional regulators of gene expression and play a pivotal role in health and disease. Exosomes are extracellular vesicles released from cells and mediate intercellular communication. Whether nut consumption modulates circulating miRNAs (c-miRNAs) transported in exosomes is poorly described. METHODS Cognitively healthy elderly subjects were randomized to either control (n = 110, abstaining from walnuts) or daily supplementation with walnuts (15% of their total energy, ≈30-60 g/day, n = 101) for 1-year. C-miRNAs were screened in exosomes isolated from 10 samples, before and after supplementation, and identified c-miRNA candidates were validated in the whole cohort. In addition, nanoparticle tracking analysis and lipidomics were assessed in pooled exosomes from the whole cohort. RESULTS Exosomal hsa-miR-32-5p and hsa-miR-29b-3p were consistently induced by walnut consumption. No major changes in exosomal lipids, nanoparticle concentration or size were found. CONCLUSION Our results provide novel evidence that certain c-miRNAs transported in exosomes are modulated by walnut consumption. The extent to which this finding contributes to the benefits of walnuts deserves further research.
Collapse
Affiliation(s)
- María-Carmen López de Las Hazas
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, IMDEA Food Institute, CEI UAM+CSIC, Ctra. De Cantoblanco 8, 28049, Madrid, Spain
| | - Judit Gil-Zamorano
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, IMDEA Food Institute, CEI UAM+CSIC, Ctra. De Cantoblanco 8, 28049, Madrid, Spain
| | - Montserrat Cofán
- Lipid Clinic, Endocrinology and Nutrition Service, Institut d´Investigacions Biomèdiques August Pi i Sunyer, 08036, Barcelona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Diana C Mantilla-Escalante
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, IMDEA Food Institute, CEI UAM+CSIC, Ctra. De Cantoblanco 8, 28049, Madrid, Spain
| | - Almudena Garcia-Ruiz
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, IMDEA Food Institute, CEI UAM+CSIC, Ctra. De Cantoblanco 8, 28049, Madrid, Spain
| | - Lorena Del Pozo-Acebo
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, IMDEA Food Institute, CEI UAM+CSIC, Ctra. De Cantoblanco 8, 28049, Madrid, Spain
| | - Oscar Pastor
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
- Servicio de Bioquímica Clínica (UCA-CCM), Hospital Ramón y Cajal-IRYCIS, 28034, Madrid, Spain
| | - María Yañez-Mo
- Department of Molecular Biology, UAM, 28049, Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBM-SO), Instituto de Investigación Sanitaria Princesa (IIS-IP), 28049, Madrid, Spain
| | - Carla Mazzeo
- Department of Molecular Biology, UAM, 28049, Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBM-SO), Instituto de Investigación Sanitaria Princesa (IIS-IP), 28049, Madrid, Spain
| | - Mercè Serra-Mir
- Lipid Clinic, Endocrinology and Nutrition Service, Institut d´Investigacions Biomèdiques August Pi i Sunyer, 08036, Barcelona, Spain
| | - Monica Doménech
- Lipid Clinic, Endocrinology and Nutrition Service, Institut d´Investigacions Biomèdiques August Pi i Sunyer, 08036, Barcelona, Spain
| | - Cinta Valls-Pedret
- Lipid Clinic, Endocrinology and Nutrition Service, Institut d´Investigacions Biomèdiques August Pi i Sunyer, 08036, Barcelona, Spain
| | - Sujatha Rajaram
- Center for Nutrition, Healthy Lifestyle and Disease Prevention, School of Public Health, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Joan Sabaté
- Center for Nutrition, Healthy Lifestyle and Disease Prevention, School of Public Health, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Emilio Ros
- Lipid Clinic, Endocrinology and Nutrition Service, Institut d´Investigacions Biomèdiques August Pi i Sunyer, 08036, Barcelona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Aleix Sala-Vila
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, 08003, Spain.
- Hospital del Mar Medical Research Institute, IMIM, Dr. Aiguader 88, 08003, Barcelona, Spain.
| | - Alberto Dávalos
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, IMDEA Food Institute, CEI UAM+CSIC, Ctra. De Cantoblanco 8, 28049, Madrid, Spain.
| |
Collapse
|
279
|
Cavallaro S, Hååg P, Viktorsson K, Krozer A, Fogel K, Lewensohn R, Linnros J, Dev A. Comparison and optimization of nanoscale extracellular vesicle imaging by scanning electron microscopy for accurate size-based profiling and morphological analysis. NANOSCALE ADVANCES 2021; 3:3053-3063. [PMID: 36133670 PMCID: PMC9419097 DOI: 10.1039/d0na00948b] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/21/2021] [Indexed: 05/05/2023]
Abstract
Nanosized extracellular vesicles (EVs) have been found to play a key role in intercellular communication, offering opportunities for both disease diagnostics and therapeutics. However, lying below the diffraction limit and also being highly heterogeneous in their size, morphology and abundance, these vesicles pose significant challenges for physical characterization. Here, we present a direct visual approach for their accurate morphological and size-based profiling by using scanning electron microscopy (SEM). To achieve that, we methodically examined various process steps and developed a protocol to improve the throughput, conformity and image quality while preserving the shape of EVs. The study was performed with small EVs (sEVs) isolated from a non-small-cell lung cancer (NSCLC) cell line as well as from human serum, and the results were compared with those obtained from nanoparticle tracking analysis (NTA). While the comparison of the sEV size distributions showed good agreement between the two methods for large sEVs (diameter > 70 nm), the microscopy based approach showed a better capacity for analyses of smaller vesicles, with higher sEV counts compared to NTA. In addition, we demonstrated the possibility of identifying non-EV particles based on size and morphological features. The study also showed process steps that can generate artifacts bearing resemblance with sEVs. The results therefore present a simple way to use a widely available microscopy tool for accurate and high throughput physical characterization of EVs.
Collapse
Affiliation(s)
- Sara Cavallaro
- Department of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology 10691 Stockholm Sweden
| | - Petra Hååg
- Department of Oncology-Pathology, Karolinska Institutet 17164 Solna Sweden
| | | | - Anatol Krozer
- Department of Smart Hardware, Division of Digital Systems, Research Institutes of Sweden AB 40014 Gothenburg Sweden
| | - Kristina Fogel
- Department of Smart Hardware, Division of Digital Systems, Research Institutes of Sweden AB 40014 Gothenburg Sweden
| | - Rolf Lewensohn
- Department of Oncology-Pathology, Karolinska Institutet 17164 Solna Sweden
- Theme Cancer, Medical Unit Head and Neck, Lung, and Skin Tumors, Thoracic Oncology Center, Karolinska University Hospital 17164 Solna Sweden
| | - Jan Linnros
- Department of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology 10691 Stockholm Sweden
| | - Apurba Dev
- Department of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology 10691 Stockholm Sweden
- Department of Electrical Engineering, The Ångström Laboratory, Uppsala University 75121 Uppsala Sweden
| |
Collapse
|
280
|
Increased Production of Outer Membrane Vesicles by Salmonella Interferes with Complement-Mediated Innate Immune Attack. mBio 2021; 12:e0086921. [PMID: 34061589 PMCID: PMC8262969 DOI: 10.1128/mbio.00869-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bacterial outer membrane vesicles (OMVs) enriched with bioactive proteins, toxins, and virulence factors play a critical role in host-pathogen and microbial interactions. The two-component system PhoP-PhoQ (PhoPQ) of Salmonella enterica orchestrates the remodeling of outer membrane lipopolysaccharide (LPS) molecules and concomitantly upregulates OMV production. In this study, we document a novel use of nanoparticle tracking analysis to determine bacterial OMV size and number. Among the PhoPQ-activated genes tested, pagC expression had the most significant effect on the upregulation of OMV production. We provide the first evidence that PhoPQ-mediated upregulation of OMV production contributes to bacterial survival by interfering with complement activation. OMVs protected bacteria in a dose-dependent manner, and bacteria were highly susceptible to complement-mediated killing in their absence. OMVs from bacteria expressing PagC bound to complement component C3b in a dose-dependent manner and inactivated it by recruiting complement inhibitor Factor H. As we also found that Factor H binds to PagC, we propose that PagC interferes with complement-mediated killing of Salmonella in the following two steps: first by engaging Factor H, and second, through the production of PagC-enriched OMVs that divert and inactivate the complement away from the bacteria. Since PhoPQ activation occurs intracellularly, the resultant increase in PagC expression and OMV production is suggested to contribute to the local and systemic spread of Salmonella released from dying host cells that supports the infection of new cells. IMPORTANCE Bacterial outer membrane vesicles (OMVs) mediate critical bacterium-bacterium and host-microbial interactions that influence pathogenesis through multiple mechanisms, including the elicitation of inflammatory responses, delivery of virulence factors, and enhancement of biofilm formation. As such, there is a growing interest in understanding the underlying mechanisms of OMV production. Recent studies have revealed that OMV biogenesis is a finely tuned physiological process that requires structural organization and selective sorting of outer membrane components into the vesicles. In Salmonella, outer membrane remodeling and OMV production are tightly regulated by its PhoPQ system. In this study, we demonstrate that PhoPQ-regulated OMV production plays a significant role in defense against host innate immune attack. PhoPQ-activated PagC expression recruits the complement inhibitor Factor H and degrades the active C3 component of complement. Our results provide valuable insight into the combination of tools and environmental signals that Salmonella employs to evade complement-mediated lysis, thereby suggesting a strong evolutionary adaptation of this facultative intracellular pathogen to protect itself during its extracellular stage in the host.
Collapse
|
281
|
Guo C, Fordjour FK, Tsai SJ, Morrell JC, Gould SJ. Choice of selectable marker affects recombinant protein expression in cells and exosomes. J Biol Chem 2021; 297:100838. [PMID: 34051235 PMCID: PMC8258971 DOI: 10.1016/j.jbc.2021.100838] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/14/2021] [Accepted: 05/25/2021] [Indexed: 11/19/2022] Open
Abstract
Transgenic mammalian cells are used for numerous research, pharmaceutical, industrial, and clinical purposes, and dominant selectable markers are often used to enable the selection of transgenic cell lines. Using HEK293 cells, we show here that the choice of selectable marker gene has a significant impact on both the level of recombinant protein expression and the cell-to-cell variability in recombinant protein expression. Specifically, we observed that cell lines generated with the NeoR or BsdR selectable markers and selected in the antibiotics G418 or blasticidin, respectively, displayed the lowest level of recombinant protein expression as well as the greatest cell-to-cell variability in transgene expression. In contrast, cell lines generated with the BleoR marker and selected in zeocin yielded cell lines that expressed the highest levels of linked recombinant protein, approximately 10-fold higher than those selected using the NeoR or BsdR markers, as well as the lowest cell-to-cell variability in recombinant protein expression. Intermediate yet still-high levels of expression were observed in cells generated with the PuroR- or HygR-based vectors and that were selected in puromycin or hygromycin, respectively. Similar results were observed in the African green monkey cell line COS7. These data indicate that each combination of selectable marker and antibiotic establishes a threshold below which no cell can survive and that these thresholds vary significantly between different selectable markers. Moreover, we show that choice of selectable marker also affects recombinant protein expression in cell-derived exosomes, consistent with the hypothesis that exosome protein budding is a stochastic rather than determinative process.
Collapse
Affiliation(s)
- Chenxu Guo
- Department of Biological Chemistry, School of Medicine, Johns Hopkins University, Baltimore Maryland, USA
| | - Francis K Fordjour
- Department of Biological Chemistry, School of Medicine, Johns Hopkins University, Baltimore Maryland, USA
| | - Shang Jui Tsai
- Department of Biological Chemistry, School of Medicine, Johns Hopkins University, Baltimore Maryland, USA
| | - James C Morrell
- Department of Biological Chemistry, School of Medicine, Johns Hopkins University, Baltimore Maryland, USA
| | - Stephen J Gould
- Department of Biological Chemistry, School of Medicine, Johns Hopkins University, Baltimore Maryland, USA.
| |
Collapse
|
282
|
Serratì S, Porcelli L, Fragassi F, Garofoli M, Di Fonte R, Fucci L, Iacobazzi RM, Palazzo A, Margheri F, Cristiani G, Albano A, De Luca R, Altomare DF, Simone M, Azzariti A. The Interaction between Reactive Peritoneal Mesothelial Cells and Tumor Cells via Extracellular Vesicles Facilitates Colorectal Cancer Dissemination. Cancers (Basel) 2021; 13:cancers13102505. [PMID: 34065529 PMCID: PMC8161093 DOI: 10.3390/cancers13102505] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 12/11/2022] Open
Abstract
Advanced colorectal cancer (CRC) is highly metastatic and often results in peritoneal dissemination. The extracellular vesicles (EVs) released by cancer cells in the microenvironment are important mediators of tumor metastasis. We investigated the contribution of EV-mediated interaction between peritoneal mesothelial cells (MCs) and CRC cells in generating a pro-metastatic environment in the peritoneal cavity. Peritoneal MCs isolated from peritoneal lavage fluids displayed high CD44 expression, substantial mesothelial-to-mesenchymal transition (MMT) and released EVs that both directed tumor invasion and caused reprogramming of secretory profiles by increasing TGF-β1 and uPA/uPAR expression and MMP-2/9 activation in tumor cells. Notably, the EVs released by tumor cells induced apoptosis by activating caspase-3, peritoneal MC senescence, and MMT, thereby augmenting the tumor-promoting potential of these cells in the peritoneal cavity. By using pantoprazole, we reduced the biogenesis of EVs and their pro-tumor functions. In conclusion, our findings provided evidence of underlying mechanisms of CRC dissemination driven by the interaction of peritoneal MCs and tumor cells via the EVs released in the peritoneal cavity, which may have important implications for the clinical management of patients.
Collapse
Affiliation(s)
- Simona Serratì
- Laboratory of Nanotechnology, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (S.S.); (A.P.)
| | - Letizia Porcelli
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, Viale O. Flacco 65, 70124 Bari, Italy; (L.P.); (M.G.); (R.D.F.); (R.M.I.)
| | - Francesco Fragassi
- Department of Surgery Oncology, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (F.F.); (R.D.L.); (D.F.A.); (M.S.)
| | - Marianna Garofoli
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, Viale O. Flacco 65, 70124 Bari, Italy; (L.P.); (M.G.); (R.D.F.); (R.M.I.)
| | - Roberta Di Fonte
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, Viale O. Flacco 65, 70124 Bari, Italy; (L.P.); (M.G.); (R.D.F.); (R.M.I.)
| | - Livia Fucci
- Pathology Department, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (L.F.); (G.C.)
| | - Rosa Maria Iacobazzi
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, Viale O. Flacco 65, 70124 Bari, Italy; (L.P.); (M.G.); (R.D.F.); (R.M.I.)
| | - Antonio Palazzo
- Laboratory of Nanotechnology, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (S.S.); (A.P.)
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy;
| | - Grazia Cristiani
- Pathology Department, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (L.F.); (G.C.)
| | - Anna Albano
- Clinical Trial Center, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy;
| | - Raffaele De Luca
- Department of Surgery Oncology, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (F.F.); (R.D.L.); (D.F.A.); (M.S.)
| | - Donato Francesco Altomare
- Department of Surgery Oncology, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (F.F.); (R.D.L.); (D.F.A.); (M.S.)
- Department of Emergency and Organ Transplantation, University Aldo Moro of Bari, 70124 Bari, Italy
| | - Michele Simone
- Department of Surgery Oncology, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (F.F.); (R.D.L.); (D.F.A.); (M.S.)
| | - Amalia Azzariti
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, Viale O. Flacco 65, 70124 Bari, Italy; (L.P.); (M.G.); (R.D.F.); (R.M.I.)
- Correspondence:
| |
Collapse
|
283
|
Chamberlain CA, Hatch M, Garrett TJ. Extracellular Vesicle Analysis by Paper Spray Ionization Mass Spectrometry. Metabolites 2021; 11:metabo11050308. [PMID: 34065030 PMCID: PMC8151837 DOI: 10.3390/metabo11050308] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/23/2021] [Accepted: 05/06/2021] [Indexed: 12/29/2022] Open
Abstract
Paper spray ionization mass spectrometry (PSI-MS) is a direct MS analysis technique with several reported bacterial metabolomics applications. As with most MS-based bacterial studies, all currently reported PSI-MS bacterial analyses have focused on the chemical signatures of the cellular unit. One dimension of the bacterial metabolome that is often lost in such analyses is the exometabolome (extracellular metabolome), including secreted metabolites, lipids, and peptides. A key component of the bacterial exometabolome that is gaining increased attention in the microbiology and biomedical communities is extracellular vesicles (EVs). These excreted structures, produced by cells in all domains of life, contain a variety of biomolecules responsible for a wide array of cellular functions, thus representing a core component of the bacterial secreted metabolome. Although previously examined using other MS approaches, no reports currently exist for a PSI-MS analysis of bacterial EVs, nor EVs from any other organism (exosomes, ectosomes, etc.). PSI-MS holds unique analytical strengths over other commonly used MS platforms and could thus provide an advantageous approach to EV metabolomics. To address this, we report a novel application representing, to our knowledge, the first PSI-MS analysis of EVs from any organism (using the human gut resident Oxalobacter formigenes as the experimental model, a bacterium whose EVs were never previously investigated). In this report, we show how we isolated and purified EVs from bacterial culture supernatant by EV-specific affinity chromatography, confirmed and characterized these vesicles by nanoparticle tracking analysis, analyzed the EV isolate by PSI-MS, and identified a panel of EV-derived metabolites, lipids, and peptides. This work serves as a pioneering study in the field of MS-based EV analysis and provides a new, rapid, sensitive, and economical approach to EV metabolomics.
Collapse
|
284
|
Porcelli L, Guida M, De Summa S, Di Fonte R, De Risi I, Garofoli M, Caputo M, Negri A, Strippoli S, Serratì S, Azzariti A. uPAR + extracellular vesicles: a robust biomarker of resistance to checkpoint inhibitor immunotherapy in metastatic melanoma patients. J Immunother Cancer 2021; 9:jitc-2021-002372. [PMID: 33972390 PMCID: PMC8112420 DOI: 10.1136/jitc-2021-002372] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2021] [Indexed: 12/23/2022] Open
Abstract
Background Emerging evidence has highlighted the importance of extracellular vesicle (EV)-based biomarkers of resistance to immunotherapy with checkpoint inhibitors in metastatic melanoma. Considering the tumor-promoting implications of urokinase-type plasminogen activator receptor (uPAR) signaling, this study aimed to assess uPAR expression in the plasma-derived EVs of patients with metastatic melanoma to determine its potential correlation with clinical outcomes. Methods Blood samples from 71 patients with metastatic melanoma were collected before initiating immunotherapy. Tumor-derived and immune cell-derived EVs were isolated and analyzed to assess the relative percentage of uPAR+ EVs. The associations between uPAR and clinical outcomes, sex, BRAF status, baseline lactate dehydrogenase levels and number of metastatic sites were assessed. Results Responders had a significantly lower percentage of tumor-derived, dendritic cell (DC)-derived and CD8+ T cell-derived uPAR +EVs at baseline than non-responders. The Kaplan-Meier survival curves for the uPAR+EV quartiles indicated that higher levels of melanoma-derived uPAR+ EVs were strongly correlated with poorer progression-free survival (p<0.0001) and overall survival (p<0.0001). We also found a statistically significant correlation between lower levels of uPAR+ EVs from both CD8+ T cells and DCs and better survival. Conclusions Our results indicate that higher levels of tumor-derived, DC-derived and CD8+ T cell-derived uPAR+ EVs in non-responders may represent a new biomarker of innate resistance to immunotherapy with checkpoint inhibitors. Moreover, uPAR+ EVs represent a new potential target for future therapeutic approaches.
Collapse
Affiliation(s)
- Letizia Porcelli
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Michele Guida
- Rare tumors and Melanoma Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Simona De Summa
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Roberta Di Fonte
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Ivana De Risi
- Rare tumors and Melanoma Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Marianna Garofoli
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Mariapia Caputo
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Antonio Negri
- Haematology Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Sabino Strippoli
- Rare tumors and Melanoma Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Simona Serratì
- Laboratory of Nanotechnology, IRCCS-Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Amalia Azzariti
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| |
Collapse
|
285
|
Zhang M, Ghosh S, Kumar M, Santiana M, Bleck CKE, Chaimongkol N, Altan-Bonnet N, Shuai D. Emerging Pathogenic Unit of Vesicle-Cloaked Murine Norovirus Clusters is Resistant to Environmental Stresses and UV 254 Disinfection. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2021; 55:6197-6205. [PMID: 33856208 DOI: 10.1021/acs.est.1c01763] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
An individual virion was long believed to act as an independent infectious unit in virology, until the recent discovery of vesicle-cloaked virus clusters which has greatly challenged this central paradigm. Vesicle-cloaked virus clusters (also known as viral vesicles) are phospholipid-bilayer encapsulated fluid sacs that contain multiple virions or multiple copies of viral genomes. Norovirus is a global leading causative agent of gastroenteritis, and the reported prevalence of vesicle-cloaked norovirus clusters in stool has raised concerns whether the current disinfection, sanitation, and hygiene practices can effectively control environmental pollution by these pathogenic units. In this study, we have demonstrated that vesicle-cloaked murine norovirus (MNV-1) clusters were highly persistent under temperature variation (i.e., freeze-thaw) and they were partially resistant to detergent decomposition. MNV-1 vesicles were 1.89-3.17-fold more infectious in vitro than their free virus counterparts. Most importantly, MNV-1 vesicles were up to 2.16-times more resistant to UV254 disinfection than free MNV-1 at a low viral load in vitro. Interestingly, with the increase of the viral load, free MNV-1 and MNV-1 vesicles showed equivalent resistance to UV254 disinfection. We show that the increased multiplicity of infection provided by vesicles is in part responsible for these attributes. Our study, for the first time, sheds light on the environmental behavior of vesicle-cloaked virus clusters as unique emerging pathogenic units. Our study highlights the need to revisit current paradigms of disinfection, sanitation, and hygiene practices for protecting public health.
Collapse
Affiliation(s)
- Mengyang Zhang
- Department of Civil and Environmental Engineering, The George Washington University, Washington, District of Columbia 20052, United States
- Laboratory of Host-Pathogen Dynamics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Sourish Ghosh
- Laboratory of Host-Pathogen Dynamics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Manish Kumar
- Laboratory of Host-Pathogen Dynamics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Marianita Santiana
- Laboratory of Host-Pathogen Dynamics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Christopher K E Bleck
- Electron Microcopy Core, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Natthawan Chaimongkol
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Nihal Altan-Bonnet
- Laboratory of Host-Pathogen Dynamics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Danmeng Shuai
- Department of Civil and Environmental Engineering, The George Washington University, Washington, District of Columbia 20052, United States
| |
Collapse
|
286
|
Yan LT, Yang ZH, Lin H, Jiang J, Jiang R. Effects of androgen on extracellular vesicles from endothelial cells in rat penile corpus cavernosum. Andrology 2021; 9:1010-1017. [PMID: 33484224 DOI: 10.1111/andr.12980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/11/2021] [Accepted: 01/20/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND The explicit mechanism of erectile dysfunction caused by low androgen status is unknown. It was reported that eNOS was expressed in extracellular vesicles (EVs). Androgen may regulate erectile function by affect the release of EVs from endothelial cells. OBJECTIVES To investigate whether androgen affects the production of EVs and nitric oxide (NO) in endothelial cells of rat penile corpus cavernosum. MATERIALS AND METHODS Endothelial cells of rat penile corpus cavernosum were isolated and purified from 6-week-old healthy male Sprague Dawley (SD) rats. Endothelial cells were treated with different concentrations of dihydrotestosterone (DHT) in a cell culture medium as follows: no-androgen group (NA group, DHT 0 nmol/L), very-low androgen group (VLA group, DHT 0.1 nmol/L), low androgen group (LA group, DHT 1 nmol/L), and physiological concentrations androgen group (PA group, DHT 10 nmol/L). After 24 h, EVs of supernatant in each group were isolated and identified. The content of EVs and NO in the supernatant and the expression of CD9, CD63, TSG101, and eNOS in EVs were detected. RESULTS Positive expression of CD9, CD63, TSG101, and eNOS was found in isolated EVs. The concentration of EVs was lower in the NA group compared with other groups (p < 0.01). The expression of eNOS and the concentration of NO was lower in the NA group than that in other groups (p < 0.05); it was lower in the VLA group than that in the LA group (p < 0.05) and lower in LA group than that in PA group (p < 0.05). When the concentration of DHT in endothelial cell culture medium ranged from 0 to 10 nmol/L, the concentration of DHT was positively correlated with the content of EVs and NO. CONCLUSION Decrease in eNOS-expressing EVs is one mechanism of NO reduction in endothelial cells of rat corpus cavernosum caused by low androgen levels.
Collapse
Affiliation(s)
- Ling-Tao Yan
- Department of Urology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Zhi-Hui Yang
- Department of Pathology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Haocheng Lin
- Department of Urology and Andrology, Peking University Third Hospital, Beijing, China
| | - Jun Jiang
- Department of Thyroid Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Rui Jiang
- Department of Urology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nephropathy Clinical Medical Research Center of Sichuan Province, Luzhou, Sichuan, China
| |
Collapse
|
287
|
Liang Y, Lehrich BM, Zheng S, Lu M. Emerging methods in biomarker identification for extracellular vesicle-based liquid biopsy. J Extracell Vesicles 2021; 10:e12090. [PMID: 34012517 PMCID: PMC8114032 DOI: 10.1002/jev2.12090] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/17/2021] [Accepted: 04/13/2021] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) are released by many cell types and distributed within various biofluids. EVs have a lipid membrane-confined structure that allows for carrying unique molecular information originating from their parent cells. The species and quantity of EV cargo molecules, including nucleic acids, proteins, lipids, and metabolites, may vary largely owing to their parent cell types and the pathophysiologic status. Such heterogeneity in EV populations provides immense challenges to researchers, yet allows for the possibility to prognosticate the pathogenesis of a particular tissue from unique molecular signatures of dispersing EVs within biofluids. However, the inherent nature of EV's small size requires advanced methods for EV purification and evaluation from the complex biofluid. Recently, the interdisciplinary significance of EV research has attracted growing interests, and the EV analytical platforms for their diagnostic prospect have markedly progressed. This review summarizes the recent advances in these EV detection techniques and methods with the intention of translating an EV-based liquid biopsy into clinical practice. This article aims to present an overview of current EV assessment techniques, with a focus on their progress and limitations, as well as an outlook on the clinical translation of an EV-based liquid biopsy that may augment current paradigms for the diagnosis, prognosis, and monitoring the response to therapy in a variety of disease settings.
Collapse
Affiliation(s)
- Yaxuan Liang
- Center for Biological Science and Technology, Advanced Institute of Natural SciencesBeijing Normal University at ZhuhaiZhuhaiChina
| | - Brandon M. Lehrich
- Medical Scientist Training ProgramUniversity of Pittsburgh School of Medicine and Carnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Siyang Zheng
- Department Biomedical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
- Department of Electrical and Computer EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Mengrou Lu
- Department Biomedical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| |
Collapse
|
288
|
Małys MS, Aigner C, Schulz SM, Schachner H, Rees AJ, Kain R. Isolation of Small Extracellular Vesicles from Human Sera. Int J Mol Sci 2021; 22:ijms22094653. [PMID: 33925027 PMCID: PMC8124960 DOI: 10.3390/ijms22094653] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 11/17/2022] Open
Abstract
Robust, well-characterized methods for purifying small extracellular vesicles (sEV) from blood are needed before their potential as disease biomarkers can be realized. Here, we compared isolation of sEV from serum by differential ultracentrifugation (DUC) and by exclusion chromatography using commercially available Exo-spin™ columns. We show that sEV can be purified by both methods but Exo-spin™ columns contain copious additional particles recorded by nanoparticle tracking analysis, invalidating its use for quantifying yields. DUC samples contained higher concentrations of exosome specific proteins CD9, CD63 and CD81 and electron microscopy confirmed that most particles in DUC preparations were sEV, whereas Exo-spin™ samples also contained copious co-purified plasma lipids. MACSPlex bead analysis identified multiple exosome surface proteins, with stronger signals in DUC samples, enabling detection of 21 of 37, compared to only 10 in Exo-spin™ samples. Nevertheless, the pattern of expression was consistent in both preparations, indicating that lipids do not interfere with bead-based technologies. Thus, both DUC and Exo-spin™ can be used to isolate sEV from human serum and what is most appropriate depends on the subsequent use of sEV. In summary, Exo-spin™ enables isolation of sEV from blood with vesicle populations similar to the ones recovered by DUC, but with lower concentrations.
Collapse
Affiliation(s)
- Małgorzata S. Małys
- Department of Pathology, Medical University Vienna, 1090 Vienna, Austria; (M.S.M.); (C.A.); (S.M.S.); (H.S.); (A.J.R.)
| | - Christof Aigner
- Department of Pathology, Medical University Vienna, 1090 Vienna, Austria; (M.S.M.); (C.A.); (S.M.S.); (H.S.); (A.J.R.)
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, 1090 Vienna, Austria
| | - Stefan M. Schulz
- Department of Pathology, Medical University Vienna, 1090 Vienna, Austria; (M.S.M.); (C.A.); (S.M.S.); (H.S.); (A.J.R.)
| | - Helga Schachner
- Department of Pathology, Medical University Vienna, 1090 Vienna, Austria; (M.S.M.); (C.A.); (S.M.S.); (H.S.); (A.J.R.)
| | - Andrew J. Rees
- Department of Pathology, Medical University Vienna, 1090 Vienna, Austria; (M.S.M.); (C.A.); (S.M.S.); (H.S.); (A.J.R.)
| | - Renate Kain
- Department of Pathology, Medical University Vienna, 1090 Vienna, Austria; (M.S.M.); (C.A.); (S.M.S.); (H.S.); (A.J.R.)
- Correspondence:
| |
Collapse
|
289
|
Aydin Y, Koksal AR, Reddy V, Lin D, Osman H, Heidari Z, Rhadhi SM, Wimley WC, Parsi MA, Dash S. Extracellular Vesicle Release Promotes Viral Replication during Persistent HCV Infection. Cells 2021; 10:984. [PMID: 33922397 PMCID: PMC8146326 DOI: 10.3390/cells10050984] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022] Open
Abstract
Hepatitis C virus (HCV) infection promotes autophagic degradation of viral replicative intermediates for sustaining replication and spread. The excessive activation of autophagy can induce cell death and terminate infection without proper regulation. A prior publication from this laboratory showed that an adaptive cellular response to HCV microbial stress inhibits autophagy through beclin 1 degradation. The mechanisms of how secretory and degradative autophagy are regulated during persistent HCV infection is unknown. This study was performed to understand the mechanisms of viral persistence in the absence of degradative autophagy, which is essential for virus survival. Using HCV infection of a CD63-green fluorescence protein (CD63-GFP), labeled stable transfected Huh-7.5 cell, we found that autophagy induction at the early stage of HCV infection increased the degradation of CD63-GFP that favored virus replication. However, the late-stage of persistent HCV infection showed impaired autophagic degradation, leading to the accumulation of CD63-GFP. We found that impaired autophagic degradation promoted the release of extracellular vesicles and exosomes. The impact of blocking the release of extracellular vesicles (EVs) on virus survival was investigated in persistently infected cells and sub-genomic replicon cells. Our study illustrates that blocking EV and exosome release severely suppresses virus replication without effecting host cell viability. Furthermore, we found that blocking EV release triggers interferon lambda 1 secretion. These findings suggest that the release of EVs is an innate immune escape mechanism that promotes persistent HCV infection. We propose that inhibition of extracellular vesicle release can be explored as a potential antiviral strategy for the treatment of HCV and other emerging RNA viruses.
Collapse
Affiliation(s)
- Yucel Aydin
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; (Y.A.); (V.R.); (D.L.); (H.O.); (S.M.R.)
| | - Ali Riza Koksal
- Department of Medicine, Division of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; (A.R.K.); (M.A.P.)
| | - Venu Reddy
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; (Y.A.); (V.R.); (D.L.); (H.O.); (S.M.R.)
| | - Dong Lin
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; (Y.A.); (V.R.); (D.L.); (H.O.); (S.M.R.)
| | - Hanadi Osman
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; (Y.A.); (V.R.); (D.L.); (H.O.); (S.M.R.)
| | - Zahra Heidari
- Department of Chemical and Biomedical Engineering, Tulane University, New Orleans, LA 70112, USA;
| | - Sadeq Mutlab Rhadhi
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; (Y.A.); (V.R.); (D.L.); (H.O.); (S.M.R.)
| | - William C Wimley
- Department of Biochemistry and Molecular Biology, Tulane University Health Sciences Center, New Orleans, LA 70112, USA;
| | - Mansour A Parsi
- Department of Medicine, Division of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; (A.R.K.); (M.A.P.)
| | - Srikanta Dash
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; (Y.A.); (V.R.); (D.L.); (H.O.); (S.M.R.)
- Department of Medicine, Division of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; (A.R.K.); (M.A.P.)
- Southeast Louisiana Veterans Health Care System, 2400 Canal Street, New Orleans, LA 70119, USA
| |
Collapse
|
290
|
Luo X, Luo SZ, Xu ZX, Zhou C, Li ZH, Zhou XY, Xu MY. Lipotoxic hepatocyte-derived exosomal miR-1297 promotes hepatic stellate cell activation through the PTEN signaling pathway in metabolic-associated fatty liver disease. World J Gastroenterol 2021; 27:1419-1434. [PMID: 33911465 PMCID: PMC8047533 DOI: 10.3748/wjg.v27.i14.1419] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 02/05/2021] [Accepted: 03/08/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Exosomes play an important role in metabolic-associated fatty liver disease (MAFLD), but the mechanism by which exosomes participate in MAFLD still remain unclear.
AIM To figure out the function of lipotoxic exosomal miR-1297 in MAFLD.
METHODS MicroRNA sequencing was used to detect differentially expressed miRNAs (DE-miR) in lipotoxic exosomes derived from primary hepatocytes. Bioinformatic tools were applied to analyze the target genes and pathways regulated by the DE-miRs. Quantitative real-time PCR (qPCR) was conducted for the verification of DE-miRs. qPCR, western blot, immunofluorescence staining and ethynyl-20-deoxyuridine assay were used to evaluate the function of lipotoxic exosomal miR-1297 on hepatic stellate cells (LX2 cells). A luciferase reporter experiment was performed to confirm the relationship of miR-1297 and its target gene PTEN.
RESULTS MicroRNA sequencing revealed that there were 61 exosomal DE-miRs (P < 0.05) with a fold-change > 2 from palmitic acid treated primary hepatocytes compared with the vehicle control group. miR-1297 was the most highly upregulated according to the microRNA sequencing. Bioinformatic tools showed a variety of target genes and pathways regulated by these DE-miRs were related to liver fibrosis. miR-1297 was overexpressed in exosomes derived from lipotoxic hepatocytes by qPCR. Fibrosis promoting genes (α-SMA, PCNA) were altered in LX2 cells after miR-1297 overexpression or miR-1297-rich lipotoxic exosome incubation via qPCR and western blot analysis. Immunofluorescence staining and ethynyl-20-deoxyuridine staining demonstrated that the activation and proliferation of LX2 cells were also promoted after the above treatment. PTEN was found to be the target gene of miR-1297 and knocking down PTEN contributed to the activation and proliferation of LX2 cells via modulating the PI3K/AKT signaling pathway.
CONCLUSION miR-1297 was overexpressed in exosomes derived from lipotoxic hepatocytes. The lipotoxic hepatocyte-derived exosomal miR-1297 could promote the activation and proliferation of hepatic stellate cells through the PTEN/PI3K/AKT signaling pathway, accelerating the progression of MAFLD.
Collapse
Affiliation(s)
- Xin Luo
- Department of Gastroenterology, Shanghai General Hospital, Shanghai 200080, China
| | - Sheng-Zheng Luo
- Department of Gastroenterology, Shanghai General Hospital, Shanghai 200080, China
| | - Zi-Xin Xu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai 200080, China
| | - Cui Zhou
- Department of Gastroenterology, Shanghai General Hospital, Shanghai 200080, China
| | - Zheng-Hong Li
- Department of Gastroenterology, Shanghai General Hospital, Shanghai 200080, China
| | - Xiao-Yan Zhou
- Department of Gastroenterology, Shanghai General Hospital, Shanghai 200080, China
| | - Ming-Yi Xu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai 200080, China
| |
Collapse
|
291
|
Zhang J, Nguyen LTH, Hickey R, Walters N, Wang X, Kwak KJ, Lee LJ, Palmer AF, Reátegui E. Immunomagnetic sequential ultrafiltration (iSUF) platform for enrichment and purification of extracellular vesicles from biofluids. Sci Rep 2021; 11:8034. [PMID: 33850163 PMCID: PMC8044115 DOI: 10.1038/s41598-021-86910-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) derived from tumor cells have the potential to provide a much-needed source of non-invasive molecular biomarkers for liquid biopsies. However, current methods for EV isolation have limited specificity towards tumor-derived EVs that limit their clinical use. Here, we present an approach called immunomagnetic sequential ultrafiltration (iSUF) that consists of sequential stages of purification and enrichment of EVs in approximately 2 h. In iSUF, EVs present in different volumes of biofluids (0.5-100 mL) can be significantly enriched (up to 1000 times), with up to 99% removal of contaminating proteins (e.g., albumin). The EV recovery rate by iSUF for cell culture media (CCM), serum, and urine corresponded to 98.0% ± 3.6%, 96.0% ± 2.0% and 94.0% ± 1.9%, respectively (p > 0.05). The final step of iSUF enables the separation of tumor-specific EVs by incorporating immunomagnetic beads to target EV subpopulations. Serum from a cohort of clinical samples from metastatic breast cancer (BC) patients and healthy donors were processed by the iSUF platform and the isolated EVs from patients showed significantly higher expression levels of BC biomarkers (i.e., HER2, CD24, and miR21).
Collapse
Affiliation(s)
- Jingjing Zhang
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Luong T H Nguyen
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Richard Hickey
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Nicole Walters
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Xinyu Wang
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Kwang Joo Kwak
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - L James Lee
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Andre F Palmer
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Eduardo Reátegui
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA.
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
292
|
Maiullari F, Chirivì M, Costantini M, Ferretti AM, Recchia S, Maiullari S, Milan M, Presutti D, Pace V, Raspa M, Scavizzi F, Massetti M, Petrella L, Fanelli M, Rizzi M, Fortunato O, Moretti F, Caradonna E, Bearzi C, Rizzi R. In vivoorganized neovascularization induced by 3D bioprinted endothelial-derived extracellular vesicles. Biofabrication 2021; 13. [PMID: 33434889 DOI: 10.1088/1758-5090/abdacf] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/12/2021] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles (EVs) have become a key tool in the biotechnological landscape due to their well-documented ability to mediate intercellular communication. This feature has been explored and is under constant investigation by researchers, who have demonstrated the important role of EVs in several research fields ranging from oncology to immunology and diagnostics to regenerative medicine. Unfortunately, there are still some limitations to overcome before clinical application, including the inability to confine the EVs to strategically defined sites of interest to avoid side effects. In this study, for the first time, EV application is supported by 3D bioprinting technology to develop a new strategy for applying the angiogenic cargo of human umbilical vein endothelial cell-derived EVs in regenerative medicine. EVs, derived from human endothelial cells and grown under different stressed conditions, were collected and used as bioadditives for the formulation of advanced bioinks. Afterin vivosubcutaneous implantation, we demonstrated that the bioprinted 3D structures, loaded with EVs, supported the formation of a new functional vasculaturein situ, consisting of blood-perfused microvessels recapitulating the printed pattern. The results obtained in this study favour the development of new therapeutic approaches for critical clinical conditions, such as the need for prompt revascularization of ischaemic tissues, which represent the fundamental substrate for advanced regenerative medicine applications.
Collapse
Affiliation(s)
- Fabio Maiullari
- Gemelli Molise SpA, Campobasso, Italy.,Istituto Nazionale Genetica Molecolare INGM 'Romeo ed Enrica Invernizzi', Milan, Italy
| | - Maila Chirivì
- Istituto Nazionale Genetica Molecolare INGM 'Romeo ed Enrica Invernizzi', Milan, Italy.,Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy
| | - Marco Costantini
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Maria Ferretti
- Institute of Chemical Sciences and Technologies "Giulio Natta", National Research Council of Italy (SCITEC-CNR), Milano, Italy
| | - Sandro Recchia
- Department of Science and High Technology, University of Insubria, Como, Italy
| | - Silvia Maiullari
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy.,Institute of Pathology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marika Milan
- Istituto Nazionale Genetica Molecolare INGM 'Romeo ed Enrica Invernizzi', Milan, Italy.,Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy
| | - Dario Presutti
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy.,Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Valentina Pace
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy.,Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Marcello Raspa
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy
| | - Ferdinando Scavizzi
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy
| | - Massimo Massetti
- Department of Cardiovascular Disease, IRCCS Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| | - Lella Petrella
- Laboratory of Molecular Oncology, Gemelli Molise SpA, Campobasso, Italy
| | - Mara Fanelli
- Laboratory of Molecular Oncology, Gemelli Molise SpA, Campobasso, Italy
| | - Marta Rizzi
- Ufficio Programmazione e Grant Office, National Research Council of Italy (UPGO-CNR), Rome, Italy
| | - Orazio Fortunato
- Tumor Genomics Unit, Department of Research, IRCCS Fondazione Istituto Nazionale dei Tumori, Milan, Italy
| | - Fabiola Moretti
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy.,IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Claudia Bearzi
- Istituto Nazionale Genetica Molecolare INGM 'Romeo ed Enrica Invernizzi', Milan, Italy.,Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy
| | - Roberto Rizzi
- Istituto Nazionale Genetica Molecolare INGM 'Romeo ed Enrica Invernizzi', Milan, Italy.,Institute of Biomedical Technologies, National Research Council of Italy (ITB-CNR), Segrate, Milan, Italy
| |
Collapse
|
293
|
Arab T, Mallick ER, Huang Y, Dong L, Liao Z, Zhao Z, Gololobova O, Smith B, Haughey NJ, Pienta KJ, Slusher BS, Tarwater PM, Tosar JP, Zivkovic AM, Vreeland WN, Paulaitis ME, Witwer KW. Characterization of extracellular vesicles and synthetic nanoparticles with four orthogonal single-particle analysis platforms. J Extracell Vesicles 2021; 10:e12079. [PMID: 33850608 PMCID: PMC8023330 DOI: 10.1002/jev2.12079] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 12/20/2022] Open
Abstract
We compared four orthogonal technologies for sizing, counting, and phenotyping of extracellular vesicles (EVs) and synthetic particles. The platforms were: single‐particle interferometric reflectance imaging sensing (SP‐IRIS) with fluorescence, nanoparticle tracking analysis (NTA) with fluorescence, microfluidic resistive pulse sensing (MRPS), and nanoflow cytometry measurement (NFCM). EVs from the human T lymphocyte line H9 (high CD81, low CD63) and the promonocytic line U937 (low CD81, high CD63) were separated from culture conditioned medium (CCM) by differential ultracentrifugation (dUC) or a combination of ultrafiltration (UF) and size exclusion chromatography (SEC) and characterized by transmission electron microscopy (TEM) and Western blot (WB). Mixtures of synthetic particles (silica and polystyrene spheres) with known sizes and/or concentrations were also tested. MRPS and NFCM returned similar particle counts, while NTA detected counts approximately one order of magnitude lower for EVs, but not for synthetic particles. SP‐IRIS events could not be used to estimate particle concentrations. For sizing, SP‐IRIS, MRPS, and NFCM returned similar size profiles, with smaller sizes predominating (per power law distribution), but with sensitivity typically dropping off below diameters of 60 nm. NTA detected a population of particles with a mode diameter greater than 100 nm. Additionally, SP‐IRIS, MRPS, and NFCM were able to identify at least three of four distinct size populations in a mixture of silica or polystyrene nanoparticles. Finally, for tetraspanin phenotyping, the SP‐IRIS platform in fluorescence mode was able to detect at least two markers on the same particle, while NFCM detected either CD81 or CD63. Based on the results of this study, we can draw conclusions about existing single‐particle analysis capabilities that may be useful for EV biomarker development and mechanistic studies.
Collapse
Affiliation(s)
- Tanina Arab
- Department of Molecular and Comparative Pathobiology Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Emily R Mallick
- Department of Molecular and Comparative Pathobiology Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Yiyao Huang
- Department of Molecular and Comparative Pathobiology Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Liang Dong
- Department of Urology Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Zhaohao Liao
- Department of Molecular and Comparative Pathobiology Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Zezhou Zhao
- Department of Molecular and Comparative Pathobiology Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Olesia Gololobova
- Department of Molecular and Comparative Pathobiology Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Barbara Smith
- Department of Cell Biology Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Norman J Haughey
- Department of Neurology Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Kenneth J Pienta
- Department of Urology Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Barbara S Slusher
- Department of Neurology Johns Hopkins University School of Medicine Baltimore Maryland USA.,Johns Hopkins Drug Discovery Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Patrick M Tarwater
- Department of Epidemiology Johns Hopkins University Bloomberg School of Public Health Baltimore Maryland USA
| | - Juan Pablo Tosar
- Faculty of Science Universidad de la República Montevideo Uruguay.,Functional Genomics Unit Institut Pasteur de Montevideo Montevideo Uruguay
| | - Angela M Zivkovic
- Department of Nutrition University of California Davis Davis California USA
| | - Wyatt N Vreeland
- Bioprocess Measurements Group National Institute of Standards and Technology Gaithersburg Maryland USA
| | - Michael E Paulaitis
- Center for Nanomedicine at the Wilmer Eye Institute Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology Johns Hopkins University School of Medicine Baltimore Maryland USA.,Department of Neurology Johns Hopkins University School of Medicine Baltimore Maryland USA.,The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease Johns Hopkins University School of Medicine Johns Hopkins Medicine and Johns Hopkins Bayview Medical Center Baltimore Maryland USA
| |
Collapse
|
294
|
Cavallaro S, Pevere F, Stridfeldt F, Görgens A, Paba C, Sahu SS, Mamand DR, Gupta D, El Andaloussi S, Linnros J, Dev A. Multiparametric Profiling of Single Nanoscale Extracellular Vesicles by Combined Atomic Force and Fluorescence Microscopy: Correlation and Heterogeneity in Their Molecular and Biophysical Features. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2008155. [PMID: 33682363 DOI: 10.1002/smll.202008155] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Indexed: 05/22/2023]
Abstract
Being a key player in intercellular communications, nanoscale extracellular vesicles (EVs) offer unique opportunities for both diagnostics and therapeutics. However, their cellular origin and functional identity remain elusive due to the high heterogeneity in their molecular and physical features. Here, for the first time, multiple EV parameters involving membrane protein composition, size and mechanical properties on single small EVs (sEVs) are simultaneously studied by combined fluorescence and atomic force microscopy. Furthermore, their correlation and heterogeneity in different cellular sources are investigated. The study, performed on sEVs derived from human embryonic kidney 293, cord blood mesenchymal stromal and human acute monocytic leukemia cell lines, identifies both common and cell line-specific sEV subpopulations bearing distinct distributions of the common tetraspanins (CD9, CD63, and CD81) and biophysical properties. Although the tetraspanin abundances of individual sEVs are independent of their sizes, the expression levels of CD9 and CD63 are strongly correlated. A sEV population co-expressing all the three tetraspanins in relatively high abundance, however, having average diameters of <100 nm and relatively low Young moduli, is also found in all cell lines. Such a multiparametric approach is expected to provide new insights regarding EV biology and functions, potentially deciphering unsolved questions in this field.
Collapse
Affiliation(s)
- Sara Cavallaro
- Department of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, Stockholm, 10691, Sweden
| | - Federico Pevere
- Department of Electrical Engineering, The Ångström Laboratory, Uppsala University, Uppsala, 75121, Sweden
| | - Fredrik Stridfeldt
- Department of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, Stockholm, 10691, Sweden
| | - André Görgens
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, 17177, Sweden
- Evox Therapeutics Limited, Oxford Science Park, Oxford, OX4 4HG, UK
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, 45141, Essen, Germany
| | | | - Siddharth S Sahu
- Department of Electrical Engineering, The Ångström Laboratory, Uppsala University, Uppsala, 75121, Sweden
| | - Doste R Mamand
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Dhanu Gupta
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, 17177, Sweden
- Evox Therapeutics Limited, Oxford Science Park, Oxford, OX4 4HG, UK
| | - Samir El Andaloussi
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, 17177, Sweden
- Evox Therapeutics Limited, Oxford Science Park, Oxford, OX4 4HG, UK
| | - Jan Linnros
- Department of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, Stockholm, 10691, Sweden
| | - Apurba Dev
- Department of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, Stockholm, 10691, Sweden
- Department of Electrical Engineering, The Ångström Laboratory, Uppsala University, Uppsala, 75121, Sweden
| |
Collapse
|
295
|
Liu M, Wang D, Gu S, Tian B, Liang J, Suo Q, Zhang Z, Yang G, Zhou Y, Li S. Micro/nano materials regulate cell morphology and intercellular communication by extracellular vesicles. Acta Biomater 2021; 124:130-138. [PMID: 33567350 DOI: 10.1016/j.actbio.2021.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EVs) have emerged as important nano-cargo carriers for cell-cell communication, yet how biophysical factors regulate EV-mediated signaling is not well understood. Here we show that microgrooves can modulate the morphology of endothelial cells (ECs), and regulate the phenotype of smooth muscle cells (SMCs) through EVs in co-culture. Elongated ECs, in comparison with polygonal ECs, increased the expression of contractile markers in SMCs. Depletion of EVs in the culture medium abolished this effect. Further analysis demonstrated that elongated ECs significantly upregulated miR-143/miR-145, leading to the increase of these microRNAs in EC-secreted EVs that were transferred to SMCs under a co-culture condition. Inhibition of EV secretion from ECs abolished the EC-SMC communication and the increased expression of SMC contractile markers. Moreover, electrospun nano-fibrous scaffolds with aligned fibers had the same effects as microgrooves to induce EC secretion of EVs to regulate SMC phenotypic marker expression. These results demonstrate that micro and nano materials can be used to engineer cell morphology and regulate EV secretion for cell-cell communication, which will have significant implications in the engineering of blood vessels and other tissues. STATEMENT OF SIGNIFICANCE: By manipulating EC morphology with micro/nano materials, we show that EV-mediated signaling can regulate SMC phenotypic marker expression. This is a very thorough and unique study to demonstrate the function of extracellular vesicles (EVs) as important nano-carriers in cell-cell communication. The originality of this study is to demonstrate that EC morphology modulates the phenotype of smooth muscle cells via extracellular vesicles enclosing miR143/miR145. These findings underscore the important role of biophysical changes in cell-cell communications, and provide a rational basis for engineering micro/nano materials to control cell-cell communications for cell and tissue engineering.
Collapse
Affiliation(s)
- Mengya Liu
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Dan Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Shuangying Gu
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Baoxiang Tian
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Jiaqi Liang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Qian Suo
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Zhijun Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Guoyuan Yang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Yue Zhou
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China.
| | - Song Li
- Department of Bioengineering and Department of Medicine, University of California, Los Angeles, CA 90095, United States
| |
Collapse
|
296
|
Human Plasma Extracellular Vesicle Isolation and Proteomic Characterization for the Optimization of Liquid Biopsy in Multiple Myeloma. Methods Mol Biol 2021; 2261:151-191. [PMID: 33420989 DOI: 10.1007/978-1-0716-1186-9_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer cells secrete membranous extracellular vesicles (EVs) which contain specific oncogenic molecular cargo (including oncoproteins, oncopeptides, and RNA) into their microenvironment and the circulation. As such, EVs including exosomes (small EVs) and microvesicles (large EVs) represent important circulating biomarkers for various diseases, including cancer and its progression. These circulating biomarkers offer a potentially minimally invasive and repeatable targets for analysis (liquid biopsy) that could aid in the diagnosis, risk stratification, and monitoring of cancer. Although their potential as cancer biomarkers has been promising, the identification and quantification of EVs in clinical samples remain challenging. Like EVs, other types of circulating biomarkers (including cell-free nucleic acids, cf-NAs; or circulating tumor cells, CTCs) may represent a complementary or alternative approach to cancer diagnosis. In the context of multiple myeloma (MM), a systemic cancer type that causes cancer cells to accumulate in the bone marrow, the specific role for EVs as biomarkers for diagnosis and monitoring remains undefined. Tumor heterogeneity along with the various subtypes of MM (such as non-secretory MM) that cannot be monitored using conventional testing (e.g. sequential serological testing and bone marrow biopsies) render liquid biopsy and circulating tumor-derived EVs a promising approach. In this protocol, we describe the isolation and purification of EVs from peripheral blood plasma (PBPL) collected from healthy donors and patients with MM for a biomarker discovery strategy. Our results demonstrate detection of circulating EVs from as little as 1 mL of MM patients' PBPL. High-resolution mass spectrometry (MS)-based proteomics promises to provide new avenues in identifying novel markers for detection, monitoring, and therapeutic intervention of disease. We describe biophysical characterization and quantitative proteomic profiling of disease-specific circulating EVs which may provide important implications for the development of cancer diagnostics in MM.
Collapse
|
297
|
Comfort N, Bloomquist TR, Shephard AP, Petty CR, Cunningham A, Hauptman M, Phipatanakul W, Baccarelli A. Isolation and characterization of extracellular vesicles in saliva of children with asthma. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2021; 2:29-48. [PMID: 34368811 PMCID: PMC8340923 DOI: 10.20517/evcna.2020.09] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/21/2021] [Accepted: 03/11/2021] [Indexed: 12/14/2022]
Abstract
AIM To confirm the presence of extracellular vesicles (EVs) in cell-free saliva (CFS) of children with asthma and describe the isolated EV population. METHODS A pooled sample of CFS EVs isolated from 180 participants using ExoQuick-TC was examined in downstream analyses. Transmission electron microscopy (TEM) was used to confirm the presence of EVs. Nanoparticle tracking analysis (NTA) and single particle interferometric reflectance imaging sensing (SP-IRIS) with fluorescence were used for sizing, counting, and phenotyping of EVs. Capillary immunoassays were used for protein quantitation. RESULTS TEM confirmed the presence of EVs of diverse sizes, indicating the prep contained a heterogeneous population of EVs. Capillary immunoassays confirmed the presence of EV-associated proteins (CD9, CD63, CD81, ICAM-1, and ANXA5) and indicated limited cellular contamination. As others have also reported, there were discrepancies in the EV sizing and enumeration across platforms. Fluorescent NTA detected particles with a mode diameter of ~90 nm, whereas SP-IRIS reported sizes of ~55-60 nm that more closely approximated the TEM results. Consistent with protein immunoassay results, SP-IRIS with fluorescence showed that the majority of these EVs were CD9- and CD63-positive, with little expression of CD81. CONCLUSION EVs from CFS can be isolated using a high-throughput method that can be scaled to large epidemiological studies. To our knowledge, we are the first to characterize CFS EVs from patients with asthma. The use of CFS EVs as potential novel biomarkers in asthma warrants further investigation and opens a new avenue of research for future studies.
Collapse
Affiliation(s)
- Nicole Comfort
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Tessa R. Bloomquist
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Alex P. Shephard
- NanoView Biosciences, Malvern Hills Science Park, Malvern, Worcestershire WR14 3SZ, UK
| | - Carter R. Petty
- Brigham and Women's Hospital, Boston, MA; Boston Children's Hospital, Clinical Research Center Boston, MA 02115, USA
| | | | - Marissa Hauptman
- Harvard Medical School, Boston, MA 02115, USA
- Division of Allergy and Immunology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Wanda Phipatanakul
- Harvard Medical School, Boston, MA 02115, USA
- Division of Allergy and Immunology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Andrea Baccarelli
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| |
Collapse
|
298
|
Comfort N, Cai K, Bloomquist TR, Strait MD, Ferrante AW, Baccarelli AA. Nanoparticle Tracking Analysis for the Quantification and Size Determination of Extracellular Vesicles. J Vis Exp 2021. [PMID: 33843938 DOI: 10.3791/62447] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The physiological and pathophysiological roles of extracellular vesicles (EVs) have become increasingly recognized, making the EV field a quickly evolving area of research. There are many different methods for EV isolation, each with distinct advantages and disadvantages that affect the downstream yield and purity of EVs. Thus, characterizing the EV prep isolated from a given source by a chosen method is important for interpretation of downstream results and comparison of results across laboratories. Various methods exist for determining the size and quantity of EVs, which can be altered by disease states or in response to external conditions. Nanoparticle tracking analysis (NTA) is one of the prominent technologies used for high-throughput analysis of individual EVs. Here, we present a detailed protocol for quantification and size determination of EVs isolated from mouse perigonadal adipose tissue and human plasma using a breakthrough technology for NTA representing major advances in the field. The results demonstrate that this method can deliver reproducible and valid total particle concentration and size distribution data for EVs isolated from different sources using different methods, as confirmed by transmission electron microscopy. The adaptation of this instrument for NTA will address the need for standardization in NTA methods to increase rigor and reproducibility in EV research.
Collapse
Affiliation(s)
- Nicole Comfort
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health;
| | - Kunheng Cai
- Department of Medicine, Naomi Berrie Diabetes Center, Columbia University
| | - Tessa R Bloomquist
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health
| | - Madeleine D Strait
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health
| | - Anthony W Ferrante
- Department of Medicine, Naomi Berrie Diabetes Center, Columbia University
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health;
| |
Collapse
|
299
|
Gómez-Serrano M, Ponath V, Preußer C, Pogge von Strandmann E. Beyond the Extracellular Vesicles: Technical Hurdles, Achieved Goals and Current Challenges When Working on Adipose Cells. Int J Mol Sci 2021; 22:ijms22073362. [PMID: 33805982 PMCID: PMC8036456 DOI: 10.3390/ijms22073362] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 12/17/2022] Open
Abstract
Adipose tissue and its crosstalk with other organs plays an essential role in the metabolic homeostasis of the entire body. Alteration of this communication (i.e., due to obesity) is related to the development of several comorbidities including type 2 diabetes, cardiovascular diseases, or cancer. Within the adipose depot, adipocytes are the main cell type and thus the main source of secreted molecules, which exert modulating effects not only at a local but also at a systemic level. Extracellular vesicles (EVs) have recently emerged as important mediators in cell–cell communication and account for part of the cellular secretome. In recent years, there has been a growing body of research on adipocyte-derived extracellular vesicles (Ad-EVs). However, there is still a lack of standardized methodological approaches, especially regarding primary adipocytes. In this review, we will provide an outline of crucial aspects when working on adipose-derived material, with a special focus on primary adipocytes. In parallel, we will point out current methodological challenges in the EV field and how they impact the transcriptomic, proteomic and functional evaluations of Ad-EVs.
Collapse
|
300
|
Prognostic plasma exosomal microRNA biomarkers in patients with substance use disorders presenting comorbid with anxiety and depression. Sci Rep 2021; 11:6271. [PMID: 33737514 PMCID: PMC7973758 DOI: 10.1038/s41598-021-84501-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/17/2021] [Indexed: 12/22/2022] Open
Abstract
Psychiatric disorders such as anxiety and depression precipitated by substance use occurred during both use and withdrawal. Exosomes play significant roles in biological functions and regulate numerous physiological and pathological processes in various diseases, in particular substance use disorders (SUDs) and other psychiatric disorders. To better understand the role of exosomal miRNAs in the pathology of symptoms of anxiety and depression in patients with SUDs, we first isolated circulating exosomes from heroin-dependent patients (HDPs) and methamphetamine-dependent patients (MDPs) and identified exosomal miRNAs that were differentially expressed between patients and healthy controls (HCs). Furthermore, the correlations between exosomal DE-miRNAs and symptoms of anxiety and depression which were measured using Hamilton-Anxiety (HAM-A)/Hamilton-Depression (HAM-D) Rating Scales in the participants. Notably, the expression level of exosomal hsa-miR-16-5p, hsa-miR-129-5p, hsa-miR-363-3p, and hsa-miR-92a-3p showed significantly negative correlations with HAM-A scores in both HDPs and MDPs. But all of the 4 DE-miRNAs lost significant correlations with HAM-D scores in HDPs. Functional annotation analyses showed that the target genes of the DE-miRNAs were mainly enriched for “synapse”, “cell adhesion”, “focal adhesion” and “MHC class II protein complex”. Our study suggests that a set of circulating exosomal miRNAs were associated with anxiety and depression in SUD patients and may have clinical utility as diagnostic and prognostic biomarkers.
Collapse
|