101
|
Nazarenko I. Extracellular Vesicles: Recent Developments in Technology and Perspectives for Cancer Liquid Biopsy. Recent Results Cancer Res 2020; 215:319-344. [PMID: 31605237 DOI: 10.1007/978-3-030-26439-0_17] [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] [Indexed: 12/13/2022]
Abstract
Extracellular micro- and nanoscale membrane vesicles produced by different cells progressively attract the attention of the scientific community. They function as mediators of intercellular communication and transport genetic material and signaling molecules between the cells. In the context of keeping homeostasis, the extracellular vesicles contribute to the regulation of various systemic and local processes. Vesicles released by the tumor and activated stromal cells exhibit multiple functions including support of tumor growth, preparation of the pre-metastatic niches, and immune suppression. Considerable progress has been made regarding the criteria of classification of the vesicles according to their origin, content, and function: Exosomes, microvesicles, also referred to as microparticles or ectosomes, and large oncosomes were defined as actively released vesicles. Additionally, apoptotic bodies represented by a highly heterogeneous population of particles produced during apoptosis, the programmed cell death, should be considered. Because the majority of isolation techniques do not allow the separation of different types of vesicles, a joined term "extracellular vesicles" (EVs) was recommended by the ISEV community for the definition of vesicles isolated from either the cell culture supernatants or the body fluids. Because EV content reflects the content of the cell of origin, multiple studies on EVs from body fluids in the context of cancer diagnosis, prediction, and prognosis were performed, actively supporting their high potential as a biomarker source. Here, we review the leading achievements in EV analysis from body fluids, defined as EV-based liquid biopsy, and provide an overview of the main EV constituents: EV surface proteins, intravesicular soluble proteins, EV RNA including mRNA and miRNA, and EV DNA as potential biomarkers. Furthermore, we discuss recent developments in technology for quantitative EV analysis in the clinical setting and future perspectives toward miniaturized high-precision liquid biopsy approaches.
Collapse
Affiliation(s)
- Irina Nazarenko
- Institute for Infection Prevention and Hospital Epidemiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany. .,German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
102
|
Ciardiello C, Migliorino R, Leone A, Budillon A. Large extracellular vesicles: Size matters in tumor progression. Cytokine Growth Factor Rev 2019; 51:69-74. [PMID: 31937439 DOI: 10.1016/j.cytogfr.2019.12.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/24/2019] [Accepted: 12/30/2019] [Indexed: 12/11/2022]
Abstract
Extracellular Vesicles (EVs) represent a heterogeneous population of particles naturally released from all cells, delimited by a lipid bilayer and able to horizontally transfer their cargos to recipient cells. These features imply the growing interest on EVs in cancer biology as biomarkers and therapeutic targets. In this review, we will highlight the specific process related to biogenesis and release of large EVs (L-EVs) derived from the plasma membrane (PM) compared to the small and well described exosomes, generated through the classical endosome-multivesicular body (MVB) pathway. The control of PM rigidity by cells depends on lipid/protein composition, cytoskeleton dynamics, cytoplasmic viscosity, ions balance, metabolic reprogramming and specific intracellular signaling pathways, all critical determinants of L-EVs biogenesis. We will focus in details on a specific class of L-EVs, named Large Oncosomes (LO), exclusively shed by cancer cells and with a size ranging from 1 μm up to 10 μm. We will examine LO specific cargos, either proteins or nucleic acids (i.e. mRNA, microRNAs, single/double-stranded DNA), as well as their functional role in cancer development and progression, also discussing the mechanisms of L-EVs internalization by recipient cells. Overall we will highlight the potential of LO as specific diagnostic/prognostic cancer biomarkers discussing the associated challenges.
Collapse
Affiliation(s)
- Chiara Ciardiello
- Experimental Pharmacology Unit, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy.
| | - Rossella Migliorino
- Experimental Pharmacology Unit, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Alessandra Leone
- Experimental Pharmacology Unit, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Alfredo Budillon
- Experimental Pharmacology Unit, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy.
| |
Collapse
|
103
|
Masvekar R, Mizrahi J, Park J, Williamson PR, Bielekova B. Quantifications of CSF Apoptotic Bodies Do Not Provide Clinical Value in Multiple Sclerosis. Front Neurol 2019; 10:1241. [PMID: 31849814 PMCID: PMC6901963 DOI: 10.3389/fneur.2019.01241] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/07/2019] [Indexed: 01/08/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS) that leads to the death of neurons and oligodendrocytes, which cannot be measured in living subjects. Physiological cellular death, otherwise known as apoptosis, progresses through a series of stages which culminates in the discharge of cellular contents into vesicles known as apoptotic bodies (ABs) or apoptosomes. These ABs can be detected in bodily fluids as Annexin-V-positive vesicles of 0.5–4.0 μm in size. In addition, the origin of these ABs might be detected by staining for cell-specific surface markers. Thus, we investigated whether quantifications of the total and CNS cell-specific ABs in the cerebrospinal fluid (CSF) of patients provided any clinical value in MS. Extracellular vesicles, from CSF of 64 prospectively-acquired subjects, were collected in a blinded fashion using ultra-centrifugation. ABs were detected by flow cytometry using bead-enabled size-gating and Annexin-V-staining. The origin of these ABs was further classified by staining the vesicles for cell-specific surface markers. Upon unblinding, we evaluated the differences between diagnostic categories and correlations with clinical measures. There were no statistically significant differences in the numbers of total or any cell-specific ABs across different disease diagnostic subgroups and no significant correlations with any of the tested clinical measures of CNS tissue destruction, disability, MS activity, and severity (i.e., rates of disability accumulation). Overlap of cell surface markers suggests inability to reliably determine origin of ABs using antibody-based flow cytometry. These negative data suggest that CNS cells in MS either die by non-apoptotic mechanisms or die in frequencies indistinguishable by current assays from apoptosis of other cells, such as immune cells performing immunosurveillance in healthy conditions.
Collapse
Affiliation(s)
- Ruturaj Masvekar
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jordan Mizrahi
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - John Park
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Peter R Williamson
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Bibiana Bielekova
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
104
|
Zhang W, Lu S, Pu D, Zhang H, Yang L, Zeng P, Su F, Chen Z, Guo M, Gu Y, Luo Y, Hu H, Lu Y, Chen F, Gao Y. Detection of fetal trisomy and single gene disease by massively parallel sequencing of extracellular vesicle DNA in maternal plasma: a proof-of-concept validation. BMC Med Genomics 2019; 12:151. [PMID: 31684971 PMCID: PMC6829814 DOI: 10.1186/s12920-019-0590-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 09/23/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND During human pregnancy, placental trophectoderm cells release extracellular vesicles (EVs) into maternal circulation. Trophoblasts also give rise to cell-free DNA (cfDNA) in maternal blood, and has been used for noninvasive prenatal screening for chromosomal aneuploidy. We intended to prove the existence of DNA in the EVs (evDNA) of maternal blood, and compared evDNA with plasma cfDNA in terms of genome distribution, fragment length, and the possibility of detecting genetic diseases. METHODS Maternal blood from 20 euploid pregnancies, 9 T21 pregnancies, 3 T18 pregnancies, 1 T13 pregnancy, and 2 pregnancies with FGFR3 mutations were obtained. EVs were separated from maternal plasma, and confirmed by transmission electronic microscopy (TEM), western blotting, and flow cytometry (FACS). evDNA was extracted and its fetal origin was confirmed by quantitative PCR (qPCR). Pair-end (PE) whole genome sequencing was performed to characterize evDNA, and the results were compared with that of cfDNA. The fetal risk of aneuploidy and monogenic diseases was analyzed using the evDNA sequencing data. RESULTS EVs separated from maternal plasma were confirmed with morphology by TEM, and protein markers of CD9, CD63, CD81 as well as the placental specific protein placental alkaline phosphatase (PLAP) were confirmed by western blotting or flow cytometry. EvDNA could be successfully extracted for qPCR and sequencing from the plasma EVs. Sequencing data showed that evDNA span on all 23 pairs of chromosomes and mitochondria, sharing a similar distribution pattern and higher GC content comparing with cfDNA. EvDNA showed shorter fragments yet lower fetal fraction than cfDNA. EvDNA could be used to correctly determine fetal gender, trisomies, and de novo FGFR3 mutations. CONCLUSIONS We proved that fetal DNA could be detected in EVs separated from maternal plasma. EvDNA shared some similar features to plasma cfDNA, and could potentially be used to detect genetic diseases in fetus.
Collapse
Affiliation(s)
- Weiting Zhang
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Sen Lu
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China
| | - Dandan Pu
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China
| | - Haiping Zhang
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China
| | - Lin Yang
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China
| | - Peng Zeng
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China
| | - Fengxia Su
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China
| | - Zhichao Chen
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Mei Guo
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Ying Gu
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China
| | - Yanmei Luo
- Prenatal Diagnosis Center, Department of Gynecology & Obstetrics, Southwest Hospital, the Third Military Medical University (Army Medical University), Chongqing, China
| | - Huamei Hu
- Prenatal Diagnosis Center, Department of Gynecology & Obstetrics, Southwest Hospital, the Third Military Medical University (Army Medical University), Chongqing, China
| | - Yanping Lu
- Department of Obstetrics and Gynecology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Fang Chen
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China.
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China.
| | - Ya Gao
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China.
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China.
| |
Collapse
|
105
|
Wu Z, Zhang Z, Xia W, Cai J, Li Y, Wu S. Extracellular vesicles in urologic malignancies-Implementations for future cancer care. Cell Prolif 2019; 52:e12659. [PMID: 31469460 PMCID: PMC6869217 DOI: 10.1111/cpr.12659] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/20/2019] [Accepted: 05/23/2019] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs), a heterogeneous group of vesicles differing in size and shape, cargo content and function, are membrane-bound and nano-sized vesicles that could be released by nearly all variations of cells. EVs have gained considerable attention in the past decades for their functions in modulating intercellular signalling and roles as potential pools for the novel diagnostic and prognostic biomarkers, as well as therapeutic targets in several cancers including urological neoplasms. In general, human and animal cells both can release distinct types of EVs, including exosomes, microvesicles, oncosomes and large oncosomes, and apoptotic bodies, while the content of EVs can be divided into proteins, lipids and nucleic acids. However, the lack of standard methods for isolation and detection platforms rein the widespread usage in clinical applications warranted furthermore investigations in the development of reliable, specific and sensitive isolation techniques. Whether and how the EVs work has become pertinent issues. With the aid of high-throughput proteomics or genomics methods, a fully understanding of contents contained in EVs from urogenital tumours, beyond all doubt, will improve our ability to identify the complex genomic alterations in the process of cancer and, in turn, contribute to detect potential therapeutic target and then provide personalization strategy for patient.
Collapse
Affiliation(s)
- Zhangsong Wu
- Medical CollegeShenzhen UniversityShenzhenChina
- Department of Urological Surgery, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
- Shenzhen Following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
| | - Zhiqiang Zhang
- Department of Urological Surgery, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
- Shenzhen Following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
| | - Wuchao Xia
- Shenzhen Following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
- Medical CollegeAnhui University of Science and TechnologyHuainanChina
| | - Jiajia Cai
- Shenzhen Following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
- Medical CollegeAnhui University of Science and TechnologyHuainanChina
| | - Yuqing Li
- Department of Urological Surgery, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
- Shenzhen Following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
| | - Song Wu
- Medical CollegeShenzhen UniversityShenzhenChina
- Department of Urological Surgery, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
- Shenzhen Following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
- Medical CollegeAnhui University of Science and TechnologyHuainanChina
- Department of Urological Surgery, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
106
|
Sheehan C, D'Souza-Schorey C. Tumor-derived extracellular vesicles: molecular parcels that enable regulation of the immune response in cancer. J Cell Sci 2019; 132:132/20/jcs235085. [PMID: 31615844 DOI: 10.1242/jcs.235085] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous collection of membrane-bound vesicles released by cells that contain bioactive cargoes including proteins, lipids and nucleic acids. Multiple subpopulations of EVs have now been recognized and these include exosomes and microvesicles. EVs have been thought to facilitate intercellular and distal communication to bring about various processes that enable tumor progression and metastases. Here, we describe the current knowledge of the functional cargo contained within EVs, with a focus on tumor microvesicles, and review the emerging theory of how EVs support immune suppression in cancer.
Collapse
Affiliation(s)
- Colin Sheehan
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556-0369, USA
| | | |
Collapse
|
107
|
Vasconcelos MH, Caires HR, Ābols A, Xavier CPR, Linē A. Extracellular vesicles as a novel source of biomarkers in liquid biopsies for monitoring cancer progression and drug resistance. Drug Resist Updat 2019; 47:100647. [PMID: 31704541 DOI: 10.1016/j.drup.2019.100647] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/09/2019] [Accepted: 10/11/2019] [Indexed: 12/23/2022]
Abstract
Cancer-derived extracellular vesicles (EVs) have been detected in the bloodstream and other biofluids of cancer patients. They carry various tumor-derived molecules such as mutated DNA and RNA fragments, oncoproteins as well as miRNA and protein signatures associated with various phenotypes. The molecular cargo of EVs partially reflects the intracellular status of their cellular origin, however various sorting mechanisms lead to the enrichment or depletion of EVs in specific nucleic acids, proteins or lipids. It is becoming increasingly clear that cancer-derived EVs act in a paracrine and systemic manner to promote cancer progression by transferring aggressive phenotypic traits and drug-resistant phenotypes to other cancer cells, modulating the anti-tumor immune response, as well as contributing to remodeling the tumor microenvironment and formation of pre-metastatic niches. These findings have raised the idea that cancer-derived EVs may serve as analytes in liquid biopsies for real-time monitoring of tumor burden and drug resistance. In this review, we have summarized recent longitudinal clinical studies describing promising EV-associated biomarkers for cancer progression and tracking cancer evolution as well as pre-clinical and clinical evidence on the relevance of EVs for monitoring the emergence or progression of drug resistance. Furthermore, we outlined the state-of-the-art in the development and commercialization of EV-based biomarkers and discussed the scientific and technological challenges that need to be met in order to translate EV research into clinically applicable tools for precision medicine.
Collapse
Affiliation(s)
- M Helena Vasconcelos
- i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy of the University of Porto, Porto, Portugal
| | - Hugo R Caires
- i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Artūrs Ābols
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Cristina P R Xavier
- i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Aija Linē
- Latvian Biomedical Research and Study Centre, Riga, Latvia; Faculty of Biology, University of Latvia, Riga, Latvia.
| |
Collapse
|
108
|
Jeong H, Shin H, Yi J, Park Y, Lee J, Gianchandani Y, Park J. Size-based analysis of extracellular vesicles using sequential transfer of an evaporating droplet. LAB ON A CHIP 2019; 19:3326-3336. [PMID: 31497821 DOI: 10.1039/c9lc00526a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
We report spatial separation of extracellular vesicle (EVs) populations based on particle size by using an approach that exploits Marangoni flow and the coffee-ring effect in microdroplets. Sequential transfer of a drying droplet progressively increases the mean size of EVs in the sample by repeated subsampling of a droplet during coffee-ring formation. This method allows size-based sorting, separation, and eventual retrieval of EVs for RNA and protein analysis. To demonstrate the biomedical relevance of this method, EVs from prostate cancer patients were analyzed; results revealed that the expression of cancer-associated genes and proteins was higher in small EVs than in large EVs. This ability to sort EVs using a combination of coffee ring with Marangoni flow and sequential droplet-transfer allows analysis of subpopulations of EVs, and will facilitate further studies of EVs.
Collapse
Affiliation(s)
| | - Hyunwoo Shin
- Mechanical Engineering, POSTECH, Republic of Korea.
| | - Johan Yi
- Mechanical Engineering, POSTECH, Republic of Korea.
| | - Yonghyun Park
- Department of Urology, Seoul St. Mary's Hospital, The Catholic University of Korea, Republic of Korea
| | - Jiyoul Lee
- Department of Urology, Seoul St. Mary's Hospital, The Catholic University of Korea, Republic of Korea
| | - Yogesh Gianchandani
- Center for Wireless Integrated MicroSensing and Systems, University of Michigan, Ann Arbor, USA.
| | - Jaesung Park
- Mechanical Engineering, POSTECH, Republic of Korea. and School of Interdisciplinary Bioscience and Bioengineering, POSTECH, Republic of Korea and Center for Wireless Integrated MicroSensing and Systems, University of Michigan, Ann Arbor, USA.
| |
Collapse
|
109
|
Qu X, Li Q, Yang J, Zhao H, Wang F, Zhang F, Zhang S, Zhang H, Wang R, Wang Q, Wang Q, Li G, Peng X, Zhou X, Hao Y, Zhu J, Xiao W. Double-Stranded DNA in Exosomes of Malignant Pleural Effusions as a Novel DNA Source for EGFR Mutation Detection in Lung Adenocarcinoma. Front Oncol 2019; 9:931. [PMID: 31608233 PMCID: PMC6773809 DOI: 10.3389/fonc.2019.00931] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/05/2019] [Indexed: 12/12/2022] Open
Abstract
Background: Exosomes are cell-derived vesicles and bear a specific set of nucleic acids including DNA (exoDNA). Thus, this study is to explore whether exoDNA in malignant pleural effusions (MPEs) could be a novel DNA source for mutation detection of epidermal growth factor receptor (EGFR). Methods: In this study, 52 lung adenocarcinoma patients were enrolled, and EGFR mutation status was detected with tumor tissues as well as cell blocks and exosomes in MPEs. The sensitivity, specificity and consistency of EGFR detection using exosomes were evaluated, compared with gene detection using tumor tissues and cell blocks. And the clinical response of patients who were detected as EGFR mutation in exosomes and treated with EGFR tyrosine kinase inhibitor (EGFR-TKI) was explored. Results: Gene detection using exosomes showed sensitivity of 100%, specificity of 96.55% and coincidence rate of 98.08% (Kappa = 0.961, P < 0.001), compared with detection using tumor tissues and cell blocks. After EGFR-TKI treatment, patients detected as EGFR mutation by exosomes showed efficacy rate of 83% and disease control rate of 100%. And patients who were detected as wild type in tumor tissues or cell blocks but EGFR mutation in exosomes turned up as PR or SD. Conclusions: These results demonstrated that exoDNA in MPEs could be used as a DNA source for EGFR detection in lung adenocarcinoma.
Collapse
Affiliation(s)
- Xueling Qu
- Department of Oncology, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Qiuwen Li
- Department of Oncology, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Jingwen Yang
- Department of Oncology, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Huixia Zhao
- Department of Oncology, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Feifei Wang
- Department of Oncology, People's Hospital of Beijing Daxing District, Capital Medical University, Beijing, China
| | - Fengyun Zhang
- Department of Oncology, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Shufang Zhang
- Department of Oncology, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - He Zhang
- Department of Oncology, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Ruliang Wang
- Department of Oncology, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Qian Wang
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qi Wang
- Department of Oncology, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Guanghui Li
- Department of Internal Medicine, Sino-Singapore Eco-City Hospital of Tianjin Medical University, Tianjin, China
| | - Xiumei Peng
- Department of Oncology, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Xuan Zhou
- Department of Oncology, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Yixin Hao
- Department of Oncology, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Jianhua Zhu
- Department of Oncology, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Wenhua Xiao
- Department of Oncology, The Fourth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
110
|
Choi D, Montermini L, Jeong H, Sharma S, Meehan B, Rak J. Mapping Subpopulations of Cancer Cell-Derived Extracellular Vesicles and Particles by Nano-Flow Cytometry. ACS NANO 2019; 13:10499-10511. [PMID: 31469961 DOI: 10.1021/acsnano.9b04480] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The elusive complexity of membranous extracellular vesicle (EV) and membrane-less extracellular particle (EP) populations released from various cellular sources contains clues as to their biological functions and diagnostic utility. In this study, we employed optimized multicolor nano-flow cytometry, structured illumination (SIM), and atomic force microscopy (AFM) to bridge sensitive detection at the single EV/EP level and high-throughput analysis of cancer cell secretomes. We applied these approaches to particles released from intact cells driven by several different transforming mechanisms or to cells under therapeutic stress imposed by pharmacological inhibition of their oncogenic drivers, such as epidermal growth factor receptor (EGFR). We demonstrate a highly heterogeneous distribution of biologically relevant elements of the EV/EP cargo, including oncoproteins (EGFR), clotting factors (tissue factor), pro-metastatic integrins (ITGA6, ITGA4), tetraspanins (CD63), and genomic DNA across the entire particulate secretome of cancer cells. We observed that targeting EGFR activity with irreversible kinase inhibitors (dacomitinib) triggers emission of DNA containing EP/EV subpopulations, including particles (chromatimeres) harboring both EGFR and DNase-resistant chromatin. While nano-flow cytometry enables quantification of these changes across the entire particular secretome, SIM reveals individual molecular topography of EV/EP subsets and AFM exposes some of their physical properties, including the presence of nanofilaments and other substructures. We describe differential uptake rates of distinct EV subsets, resulting in preferential internalization of exosome-like small EVs by cancer cells to the exclusion of larger EVs. Thus, our study illustrates the potential of nano-flow cytometry coupled with high-resolution microscopy to explore the cancer-related EV/EP landscape.
Collapse
Affiliation(s)
- Dongsic Choi
- Research Institute of the McGill University Health Centre, Glen Site , McGill University , Montreal , Quebec H4A 3J1 , Canada
| | - Laura Montermini
- Research Institute of the McGill University Health Centre, Glen Site , McGill University , Montreal , Quebec H4A 3J1 , Canada
| | - Hyeonju Jeong
- Research Institute of the McGill University Health Centre, Glen Site , McGill University , Montreal , Quebec H4A 3J1 , Canada
| | - Shivani Sharma
- Department of Pathology & Laboratory Medicine and California Nanosystems Institute , University of California at Los Angeles , Los Angeles , California 90095 , United States
| | - Brian Meehan
- Research Institute of the McGill University Health Centre, Glen Site , McGill University , Montreal , Quebec H4A 3J1 , Canada
| | - Janusz Rak
- Research Institute of the McGill University Health Centre, Glen Site , McGill University , Montreal , Quebec H4A 3J1 , Canada
| |
Collapse
|
111
|
Sharma A, Johnson A. Exosome DNA: Critical regulator of tumor immunity and a diagnostic biomarker. J Cell Physiol 2019; 235:1921-1932. [DOI: 10.1002/jcp.29153] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/26/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Aman Sharma
- ExoCan Healthcare Technologies Ltd, Lab 4 400 NCL Innovation Park Pune India
| | - Abiya Johnson
- ExoCan Healthcare Technologies Ltd, Lab 4 400 NCL Innovation Park Pune India
| |
Collapse
|
112
|
Lázaro-Ibáñez E, Lässer C, Shelke GV, Crescitelli R, Jang SC, Cvjetkovic A, García-Rodríguez A, Lötvall J. DNA analysis of low- and high-density fractions defines heterogeneous subpopulations of small extracellular vesicles based on their DNA cargo and topology. J Extracell Vesicles 2019; 8:1656993. [PMID: 31497265 PMCID: PMC6719264 DOI: 10.1080/20013078.2019.1656993] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 08/12/2019] [Accepted: 08/12/2019] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles have the capacity to transfer lipids, proteins, and nucleic acids between cells, thereby influencing the recipient cell's phenotype. While the role of RNAs in EVs has been extensively studied, the function of DNA remains elusive. Here, we distinguished novel heterogeneous subpopulations of small extracellular vesicles (sEVs) based on their DNA content and topology. Low- and high-density sEV subsets from a human mast cell line (HMC-1) and an erythroleukemic cell line (TF-1) were separated using high-resolution iodixanol density gradients to discriminate the nature of the DNA cargo of the sEVs. Paired comparisons of the sEV-associated DNA and RNA molecules showed that RNA was more abundant than DNA and that most of the DNA was present in the high-density fractions, demonstrating that sEV subpopulations have different DNA content. DNA was predominately localised on the outside or surface of sEVs, with only a small portion being protected from enzymatic degradation. Whole-genome sequencing identified DNA fragments spanning all chromosomes and mitochondrial DNA when sEVs were analysed in bulk. Our work contributes to the understanding of how DNA is associated with sEVs and thus provides direction for distinguishing subtypes of EVs based on their DNA cargo and topology.
Collapse
Affiliation(s)
- Elisa Lázaro-Ibáñez
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Cecilia Lässer
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Ganesh Vilas Shelke
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Rossella Crescitelli
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Su Chul Jang
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Aleksander Cvjetkovic
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Anaís García-Rodríguez
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
113
|
Gruzdev SK, Yakovlev AA, Druzhkova TA, Guekht AB, Gulyaeva NV. The Missing Link: How Exosomes and miRNAs can Help in Bridging Psychiatry and Molecular Biology in the Context of Depression, Bipolar Disorder and Schizophrenia. Cell Mol Neurobiol 2019; 39:729-750. [PMID: 31089834 DOI: 10.1007/s10571-019-00684-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/03/2019] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) only recently have been recognized as promising molecules for both fundamental and clinical neuroscience. We provide a literature review of miRNA biomarker studies in three most prominent psychiatric disorders (depression, bipolar disorder and schizophrenia) with the particular focus on depression due to its social and healthcare importance. Our search resulted in 191 unique miRNAs across 35 human studies measuring miRNA levels in blood, serum or plasma. 30 miRNAs replicated in more than one study. Most miRNAs targeted neuroplasticity and neurodevelopment pathways. Various limitations do not allow us to make firm conclusions on clinical potential of studied miRNAs. Based on our results we discuss the rationale for future research investigations of exosomal mechanisms to overcome methodological caveats both in studying etiology and pathogenesis, and providing an objective back-up for clinical decisions.
Collapse
Affiliation(s)
- S K Gruzdev
- Institute of Medicine, RUDN University, Miklukho-Maklaya Str. 6, Moscow, Russia, 117198.
| | - A A Yakovlev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerova Str., 5A, Moscow, Russia, 117485
- Moscow Research & Clinical Center for Neuropsychiatry, Moscow Healthcare Department, Donskaya Str., 43, Moscow, Russia, 115419
| | - T A Druzhkova
- Moscow Research & Clinical Center for Neuropsychiatry, Moscow Healthcare Department, Donskaya Str., 43, Moscow, Russia, 115419
| | - A B Guekht
- Moscow Research & Clinical Center for Neuropsychiatry, Moscow Healthcare Department, Donskaya Str., 43, Moscow, Russia, 115419
- Russian National Research Medical University, Ostrovitianov Str. 1, Moscow, Russia, 117997
| | - N V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerova Str., 5A, Moscow, Russia, 117485
- Moscow Research & Clinical Center for Neuropsychiatry, Moscow Healthcare Department, Donskaya Str., 43, Moscow, Russia, 115419
| |
Collapse
|
114
|
Wan Y, Maurer M, He HZ, Xia YQ, Hao SJ, Zhang WL, Yee NS, Zheng SY. Enrichment of extracellular vesicles with lipid nanoprobe functionalized nanostructured silica. LAB ON A CHIP 2019; 19:2346-2355. [PMID: 31232418 PMCID: PMC6669184 DOI: 10.1039/c8lc01359d] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Nanoscale extracellular vesicles (nEVs) have recently demonstrated potential value in cancer diagnostics and treatment monitoring, but translation has been limited by technical challenges in nEV isolation. Thus, we have developed a one-step nEV isolation platform that utilizes nEV size-matched silica nanostructures and a surface-conjugated lipid nanoprobe with an integrated microfluidic mixer. The reported platform has 28.8% capture efficiency from pancreatic cancer plasma and can sufficiently enrich nEVs for simpler positive identification of point mutations, particularly KRAS, in nEV DNA from the plasma of pancreatic cancer patients.
Collapse
Affiliation(s)
- Yuan Wan
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Materials Research Institute, The Pennsylvania State University, University Park, PA 16802, USA
| | - Mackenzie Maurer
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Materials Research Institute, The Pennsylvania State University, University Park, PA 16802, USA
| | - Hong-Zhang He
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Materials Research Institute, The Pennsylvania State University, University Park, PA 16802, USA
| | - Yi-Qiu Xia
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Materials Research Institute, The Pennsylvania State University, University Park, PA 16802, USA
| | - Si-Jie Hao
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Materials Research Institute, The Pennsylvania State University, University Park, PA 16802, USA
| | - Wen-Long Zhang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Materials Research Institute, The Pennsylvania State University, University Park, PA 16802, USA
| | - Nelson S. Yee
- Department of Medicine, Hematology/Oncology, Penn State Cancer Institute, Hershey, PA 17033, USA
| | - Si-Yang Zheng
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Materials Research Institute, The Pennsylvania State University, University Park, PA 16802, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, Univeristy Park, PA 16802, USA
- Department of Electrical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
115
|
Chiabotto G, Gai C, Deregibus MC, Camussi G. Salivary Extracellular Vesicle-Associated exRNA as Cancer Biomarker. Cancers (Basel) 2019; 11:cancers11070891. [PMID: 31247906 PMCID: PMC6679099 DOI: 10.3390/cancers11070891] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/11/2019] [Accepted: 06/22/2019] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) secreted in biological fluids contain several transcripts of the cell of origin, which may modify the functions and phenotype of proximal and distant cells. Cancer-derived EVs may promote a favorable microenvironment for cancer growth and invasion by acting on stroma and endothelial cells and may favor metastasis formation. The transcripts contained in cancer EVs may be exploited as biomarkers. Protein and extracellular RNA (exRNA) profiling in patient bio-fluids, such as blood and urine, was performed to identify molecular features with potential diagnostic and prognostic values. EVs are concentrated in saliva, and salivary EVs are particularly enriched in exRNAs. Several studies were focused on salivary EVs for the detection of biomarkers either of non-oral or oral cancers. The present paper provides an overview of the available studies on the diagnostic potential of exRNA profiling in salivary EVs.
Collapse
Affiliation(s)
- Giulia Chiabotto
- Department of Medical Sciences, University of Torino, Torino 10126, Italy.
| | - Chiara Gai
- Department of Medical Sciences, University of Torino, Torino 10126, Italy.
| | - Maria Chiara Deregibus
- i3T Business Incubator and Technology Transfer, University of Torino, Torino 10126, Italy.
| | - Giovanni Camussi
- Department of Medical Sciences, University of Torino, Torino 10126, Italy.
| |
Collapse
|
116
|
Konečná B, Tóthová Ľ, Repiská G. Exosomes-Associated DNA-New Marker in Pregnancy Complications? Int J Mol Sci 2019; 20:ijms20122890. [PMID: 31200554 PMCID: PMC6627934 DOI: 10.3390/ijms20122890] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/09/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022] Open
Abstract
Despite a large number of studies, the etiology of pregnancy complications remains unknown. The involvement of cell-free DNA or fetal cell-free DNA in the pathogenesis of pregnancy complications is currently being hypothesized. Cell-free DNA occurs in different forms-free; part of neutrophil extracellular traps; or as recently discovered, carried by extracellular vesicles. Cell-free DNA is believed to activate an inflammatory pathway, which could possibly cause pregnancy complications. It could be hypothesized that DNA in its free form could be easily degraded by nucleases to prevent the inflammatory activation. However, recently, there has been a growing interest in the role of exosomes, potential protectors of cell-free DNA, in pregnancy complications. Most of the interest from recent years is directed towards the micro RNA carried by exosomes. However, exosome-associated DNA in relation to pregnancy complications has not been truly studied yet. DNA, as an important cargo of exosomes, has been so far studied mostly in cancer research. This review collects all the known information on the topic of not only exosome-associated DNA but also some information on vesicles-associated DNA and the studies regarding the role of exosomes in pregnancy complications from recent years. It also suggests possible analysis of exosome-associated DNA in pregnancy from plasma and emphasizes the importance of such analysis for future investigations of pregnancy complications. A major obstacle to the advancement in this field is the proper uniformed technique for exosomes isolation. Similarly, the sensitivity of methods analyzing a small fraction of DNA, potentially fetal DNA, carried by exosomes is variable.
Collapse
Affiliation(s)
- Barbora Konečná
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava 81108, Slovakia.
| | - Ľubomíra Tóthová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava 81108, Slovakia.
| | - Gabriela Repiská
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava 81372, Slovakia.
| |
Collapse
|
117
|
Lu YT, Delijani K, Mecum A, Goldkorn A. Current status of liquid biopsies for the detection and management of prostate cancer. Cancer Manag Res 2019; 11:5271-5291. [PMID: 31239778 PMCID: PMC6559244 DOI: 10.2147/cmar.s170380] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/18/2019] [Indexed: 12/14/2022] Open
Abstract
In recent years, new therapeutic options have become available for prostate cancer (PC) patients, generating an urgent need for better biomarkers to guide the choice of therapy and monitor treatment response. Liquid biopsies, including circulating tumor cells (CTCs), circulating nucleic acids, and exosomes, have been developed as minimally invasive assays allowing oncologists to monitor PC patients with real-time cellular or molecular information. While CTC counts remain the most extensively validated prognostic biomarker to monitor treatment response, recent advances demonstrate that CTC morphology and androgen receptor characterization can provide additional information to guide the choice of treatment. Characterization of cell-free DNA (cfDNA) is another rapidly emerging field with novel technologies capable of monitoring the evolution of treatment relevant alterations such as those in DNA damage repair genes for poly (ADP-ribose) polymerase (PARP) inhibition. In addition, several new liquid biopsy fields are emerging, including the characterization of heterogeneity, CTC RNA sequencing, the culture and xenografting of CTCs, and the characterization of extracellular vesicles (EVs) and circulating microRNAs. This review describes the clinical utilization of liquid biopsies in the management of PC patients and emerging liquid biopsy technologies with the potential to advance personalized cancer therapy.
Collapse
Affiliation(s)
- Yi-Tsung Lu
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Kevin Delijani
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Andrew Mecum
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Amir Goldkorn
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
118
|
Poon IKH, Parkes MAF, Jiang L, Atkin-Smith GK, Tixeira R, Gregory CD, Ozkocak DC, Rutter SF, Caruso S, Santavanond JP, Paone S, Shi B, Hodge AL, Hulett MD, Chow JDY, Phan TK, Baxter AA. Moving beyond size and phosphatidylserine exposure: evidence for a diversity of apoptotic cell-derived extracellular vesicles in vitro. J Extracell Vesicles 2019; 8:1608786. [PMID: 31069027 PMCID: PMC6493268 DOI: 10.1080/20013078.2019.1608786] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 12/16/2022] Open
Abstract
Apoptosis is a form of programmed cell death that occurs throughout life as part of normal development as well as pathologic processes including chronic inflammation and infection. Although the death of a cell is often considered as the only biological outcome of a cell committed to apoptosis, it is becoming increasingly clear that the dying cell can actively communicate with other cells via soluble factors as well as membrane-bound extracellular vesicles (EVs) to regulate processes including cell clearance, immunity and tissue repair. Compared to EVs generated from viable cells such as exosomes and microvesicles, apoptotic cell-derived EVs (ApoEVs) are less well defined and the basic criteria for ApoEV characterization have not been established in the field. In this study, we will examine the current understanding of ApoEVs, in particular, the ApoEV subtype called apoptotic bodies (ApoBDs). We described that a subset of ApoBDs can be larger than 5 μm and smaller than 1 μm based on flow cytometry and live time-lapse microscopy analysis, respectively. We also described that a subset of ApoBDs can expose a relatively low level of phosphatidylserine on its surface based on annexin A5 staining. Furthermore, we characterized the presence of caspase-cleaved proteins (in particular plasma membrane-associated or cytoplasmic proteins) in samples enriched in ApoBDs. Lastly, using a combination of biochemical-, live imaging- and flow cytometry-based approaches, we characterized the progressive lysis of ApoBDs. Taken together, these results extended our understanding of ApoBDs.
Collapse
Affiliation(s)
- Ivan K H Poon
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Michael A F Parkes
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Lanzhou Jiang
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Georgia K Atkin-Smith
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Rochelle Tixeira
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Christopher D Gregory
- MRC Centre for inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Dilara C Ozkocak
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Stephanie F Rutter
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Sarah Caruso
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Jascinta P Santavanond
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Stephanie Paone
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Bo Shi
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Amy L Hodge
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Mark D Hulett
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Jenny D Y Chow
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Thanh Kha Phan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Amy A Baxter
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| |
Collapse
|
119
|
Repiská G, Konečná B, Shelke GV, Lässer C, Vlková BI, Minárik G. Is the DNA of placental origin packaged in exosomes isolated from plasma and serum of pregnant women? Clin Chem Lab Med 2019; 56:e150-e153. [PMID: 29306910 DOI: 10.1515/cclm-2017-0560] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 12/06/2017] [Indexed: 11/15/2022]
Affiliation(s)
- Gabriela Repiská
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 2, 813 72 Bratislava, Slovakia
| | - Barbora Konečná
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Ganesh V Shelke
- Krefting Research Centre, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Lässer
- Krefting Research Centre, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Barbora Izrael Vlková
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Gabriel Minárik
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
120
|
Virzì G, Clementi A, Battaglia G, Ronco C. Multi-Omics Approach: New Potential Key Mechanisms Implicated in Cardiorenal Syndromes. Cardiorenal Med 2019; 9:201-211. [DOI: 10.1159/000497748] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/31/2019] [Indexed: 11/19/2022] Open
Abstract
Cardiorenal syndromes (CRS) include a scenario of clinical interactions characterized by the heart and kidney dysfunction. The crosstalk between cardiac and renal systems is clearly evidenced but not completely understood. Multi-factorial mechanisms leading to CRS do not involve only hemodynamic parameters. In fact, in recent works on organ crosstalk endothelial injury, the alteration of normal immunologic balance, cell death, inflammatory cascades, cell adhesion molecules, cytokine and chemokine overexpression, neutrophil migration, leukocyte trafficking, caspase-mediated induction of apoptotic mechanisms and oxidative stress has been demonstrated to induce distant organ dysfunction. Furthermore, new alternative mechanisms using the multi-omics approach may be implicated in the pathogenesis of cardiorenal crosstalk. The study of “omics” modifications in the setting of cardiovascular and renal disease represents an emerging area of research. Over the last years, indeed, many studies have elucidated the exact mechanisms involved in gene expression and regulation, cellular communication and organ crosstalk. In this review, we analyze epigenetics, gene expression, small non-coding RNAs, extracellular vesicles, proteomics, and metabolomics in the setting of CRS.
Collapse
|
121
|
Roy S, Lin HY, Chou CY, Huang CH, Small J, Sadik N, Ayinon CM, Lansbury E, Cruz L, Yekula A, Jones PS, Balaj L, Carter BS. Navigating the Landscape of Tumor Extracellular Vesicle Heterogeneity. Int J Mol Sci 2019; 20:ijms20061349. [PMID: 30889795 PMCID: PMC6471355 DOI: 10.3390/ijms20061349] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/08/2019] [Accepted: 03/08/2019] [Indexed: 01/01/2023] Open
Abstract
The last decade has seen a rapid expansion of interest in extracellular vesicles (EVs) released by cells and proposed to mediate intercellular communication in physiological and pathological conditions. Considering that the genetic content of EVs reflects that of their respective parent cell, many researchers have proposed EVs as a source of biomarkers in various diseases. So far, the question of heterogeneity in given EV samples is rarely addressed at the experimental level. Because of their relatively small size, EVs are difficult to reliably isolate and detect within a given sample. Consequently, standardized protocols that have been optimized for accurate characterization of EVs are lacking despite recent advancements in the field. Continuous improvements in pre-analytical parameters permit more efficient assessment of EVs, however, methods to more objectively distinguish EVs from background, and to interpret multiple single-EV parameters are lacking. Here, we review EV heterogeneity according to their origin, mode of release, membrane composition, organelle and biochemical content, and other factors. In doing so, we also provide an overview of currently available and potentially applicable methods for single EV analysis. Finally, we examine the latest findings from experiments that have analyzed the issue at the single EV level and discuss potential implications.
Collapse
Affiliation(s)
- Sabrina Roy
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Hsing-Ying Lin
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Chung-Yu Chou
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City 32001, Taiwan.
| | - Chen-Han Huang
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City 32001, Taiwan.
| | - Julia Small
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Noah Sadik
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
- Department of Biomedical Engineering, Columbia University, New York City, NY 10027, USA.
| | - Caroline M Ayinon
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Elizabeth Lansbury
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Lilian Cruz
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Anudeep Yekula
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Pamela S Jones
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Leonora Balaj
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Bob S Carter
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
122
|
Abstract
Extracellular vesicles (EVs) are important mediators of intercellular communication in cancer and in normal tissues. EVs transfer biologically active molecules from the cell of origin to recipient cells. This review summarizes the studies on EVs derived from renal cell carcinoma and from a subpopulation of CD105-positive renal cancer stem cells. While EVs from renal cell carcinoma show mild biological activity, EVs from renal cancer stem cells enhance tumor angiogenesis and metastasis formation. The effect is probably due to the transfer of proangiogenic RNA cargo to endothelial cells, which acquire an activated angiogenic phenotype. In vivo, treatment with EVs favors the formation of a premetastatic niche in the lungs. Moreover, EVs derived from renal cancer stem cells modify gene expression in mesenchymal stromal cells, enhancing the expression of genes involved in matrix remodeling, cell migration, and tumor growth. Mesenchymal stromal cells preconditioned with tumor EVs and then coinjected in vivo with renal cancer cells support tumor growth and vessel formation. Finally, tumor EVs promote tumor immune escape by inhibiting the differentiation process of dendritic cells and the activation of T cells. Thus, tumor-derived EVs act on the microenvironment favoring tumor aggressiveness, may contribute to angiogenesis through both direct and indirect mechanisms and are involved in tumor immune escape. Membrane-bound packages called extracellular vesicles (EVs) released by kidney cancer stem cells can make tumors more aggressive, promote the onset of cancer at other sites, and help tumors escape the anti-cancer immune response. Giovanni Camussi and colleagues at the University of Turin, Italy, review understanding of EVs from kidney cancer cells. EVs from cancer stem cells are especially effective in promoting cancer, unlike those from mature cancer cells. This is partly due to their ability to promote the formation of new blood vessels to sustain tumor growth. Some of the vesicles’ effects are mediated by transferring small molecules of ribonucleic acid (RNA) into other cells. These RNAs can regulate the activity of specific genes, promoting cancer. Studying patients’ EVs may assist cancer diagnosis and help predict the likely progression of the disease.
Collapse
|
123
|
Gézsi A, Kovács Á, Visnovitz T, Buzás EI. Systems biology approaches to investigating the roles of extracellular vesicles in human diseases. Exp Mol Med 2019; 51:1-11. [PMID: 30872567 PMCID: PMC6418293 DOI: 10.1038/s12276-019-0226-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are membrane-enclosed structures secreted by cells. In the past decade, EVs have attracted substantial attention as carriers of complex intercellular information. They have been implicated in a wide variety of biological processes in health and disease. They are also considered to hold promise for future diagnostics and therapy. EVs are characterized by a previously underappreciated heterogeneity. The heterogeneity and molecular complexity of EVs necessitates high-throughput analytical platforms for detailed analysis. Recently, mass spectrometry, next-generation sequencing and bioinformatics tools have enabled detailed proteomic, transcriptomic, glycomic, lipidomic, metabolomic, and genomic analyses of EVs. Here, we provide an overview of systems biology experiments performed in the field of EVs. Furthermore, we provide examples of how in silico systems biology approaches can be used to identify correlations between genes involved in EV biogenesis and human diseases. Using a knowledge fusion system, we investigated whether certain groups of proteins implicated in the biogenesis/release of EVs were associated with diseases and phenotypes. Furthermore, we investigated whether these proteins were enriched in publicly available transcriptomic datasets using gene set enrichment analysis methods. We found associations between key EV biogenesis proteins and numerous diseases, which further emphasizes the key role of EVs in human health and disease.
Collapse
Affiliation(s)
- András Gézsi
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- MTA-SE Immune-Proteogenomics Extracellular Vesicle Research Group, Budapest, Hungary
- Department of Measurement and Information Systems, Budapest University of Technology and Economics, Budapest, Hungary
| | - Árpád Kovács
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Tamás Visnovitz
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Edit I Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary.
- MTA-SE Immune-Proteogenomics Extracellular Vesicle Research Group, Budapest, Hungary.
| |
Collapse
|
124
|
De Palma G, Di Lorenzo VF, Krol S, Paradiso AV. Urinary exosomal shuttle RNA: Promising cancer diagnosis biomarkers of lower urinary tract. Int J Biol Markers 2019; 34:101-107. [PMID: 30862241 DOI: 10.1177/1724600819827023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Prostate and bladder cancers continue to be the first and fourth most common cancers in men worldwide; thus there is an urgent need for more accurate biomarkers that can detect these types of cancer in a non-invasive way. Liquid biopsy is a new non-invasive tool for diagnosis and with a virtually unlimited supply urine is even more attractive resource since urinary exosomes have been discovered to contain RNAs that are hallmarks of cancer. It is challenging to assay those secreting lower amounts of molecules. METHODS This review, based on articles identified through a PubMed/MEDLINE search, comprehensively summarizes state of the art approaches used in the discovery and validation of exosomal RNA biomarkers purified from the urine for lower urinary tract cancer. RESULTS The combination of PCA3 and ERG has shown a relatively good improvement in diagnostic performance; examples of other potential biomarkers and the methods utilized in their discovery are also discussed in this review. CONCLUSIONS Of these last markers, to date there are still few data to implement these for routine diagnosis.
Collapse
Affiliation(s)
- Giuseppe De Palma
- 1 Institutional BioBank, Experimental Oncology and Biobank Management Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italia
| | | | - Silke Krol
- 3 Translational Nanotechnology Laboratory, Experimental Oncology and Biobank Management Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italia
| | - Angelo Virgilio Paradiso
- 1 Institutional BioBank, Experimental Oncology and Biobank Management Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italia
| |
Collapse
|
125
|
Davis CN, Phillips H, Tomes JJ, Swain MT, Wilkinson TJ, Brophy PM, Morphew RM. The importance of extracellular vesicle purification for downstream analysis: A comparison of differential centrifugation and size exclusion chromatography for helminth pathogens. PLoS Negl Trop Dis 2019; 13:e0007191. [PMID: 30811394 PMCID: PMC6411213 DOI: 10.1371/journal.pntd.0007191] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 03/11/2019] [Accepted: 01/27/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Robust protocols for the isolation of extracellular vesicles (EVs) from the rest of their excretory-secretory products are necessary for downstream studies and application development. The most widely used purification method of EVs for helminth pathogens is currently differential centrifugation (DC). In contrast, size exclusion chromatography (SEC) has been included in the purification pipeline for EVs from other pathogens, highlighting there is not an agreed research community 'gold standard' for EV isolation. In this case study, Fasciola hepatica from natural populations were cultured in order to collect EVs from culture media and evaluate a SEC or DC approach to pathogen helminth EV purification. METHODOLOGY/PRINCIPAL FINDINGS Transmission electron and atomic force microscopy demonstrated that EVs prepared by SEC were both smaller in size and less diverse than EV resolved by DC. Protein quantification and Western blotting further demonstrated that SEC purification realised a higher EV purity to free excretory-secretory protein (ESP) yield ratio compared to DC approaches as evident by the reduction of soluble free cathepsin L proteases in SEC EV preparations. Proteomic analysis further highlighted DC contamination from ESP as shown by an increased diversity of protein identifications and unique peptide hits in DC EVs as compared to SEC EVs. In addition, SEC purified EVs contained less tegumental based proteins than DC purified EVs. CONCLUSIONS/SIGNIFICANCE The data suggests that DC and SEC purification methods do not isolate equivalent EV population profiles and caution should be taken in the choice of EV purification utilised, with certain protocols for DC preparations including more free ES proteins and tegumental artefacts. We propose that SEC methods should be used for EV purification prior to downstream studies.
Collapse
Affiliation(s)
- Chelsea N. Davis
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| | - Helen Phillips
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| | - John J. Tomes
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| | - Martin T. Swain
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| | - Toby J. Wilkinson
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| | - Peter M. Brophy
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| | - Russell M. Morphew
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| |
Collapse
|
126
|
Gámez-Valero A, Beyer K, Borràs FE. Extracellular vesicles, new actors in the search for biomarkers of dementias. Neurobiol Aging 2019; 74:15-20. [DOI: 10.1016/j.neurobiolaging.2018.10.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 09/14/2018] [Accepted: 10/04/2018] [Indexed: 02/07/2023]
|
127
|
Choi D, Spinelli C, Montermini L, Rak J. Oncogenic Regulation of Extracellular Vesicle Proteome and Heterogeneity. Proteomics 2019; 19:e1800169. [PMID: 30561828 DOI: 10.1002/pmic.201800169] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/05/2018] [Indexed: 12/12/2022]
Abstract
Mutational and epigenetic driver events profoundly alter intercellular communication pathways in cancer. This effect includes deregulated release, molecular composition, and biological activity of extracellular vesicles (EVs), membranous cellular fragments ranging from a few microns to less than 100 nm in diameter and filled with bioactive molecular cargo (proteins, lipids, and nucleic acids). While EVs are usually classified on the basis of their physical properties and biogenetic mechanisms, recent analyses of their proteome suggest a larger than expected molecular diversity, a notion that is also supported by multicolour nano-flow cytometry and other emerging technology platforms designed to analyze single EVs. Both protein composition and EV diversity are markedly altered by oncogenic transformation, epithelial to mesenchymal transition, and differentiation of cancer stem cells. Interestingly, only a subset of EVs released from mutant cells may carry oncogenic proteins (e.g., EGFRvIII), hence, these EVs are often referred to as "oncosomes". Indeed, oncogenic transformation alters the repertoire of EV-associated proteins, increases the presence of pro-invasive cargo, and alters the composition of distinct EV populations. Molecular profiling of single EVs may reveal a more intricate effect of transforming events on the architecture of EV populations in cancer and shed new light on their biological role and diagnostic utility.
Collapse
Affiliation(s)
- Dongsic Choi
- Research Institute, Health Centre, Glen Site, McGill University, Montreal, Quebec, H4A 3J1, Canada
| | - Cristiana Spinelli
- Research Institute, Health Centre, Glen Site, McGill University, Montreal, Quebec, H4A 3J1, Canada
| | - Laura Montermini
- Research Institute, Health Centre, Glen Site, McGill University, Montreal, Quebec, H4A 3J1, Canada
| | - Janusz Rak
- Research Institute, Health Centre, Glen Site, McGill University, Montreal, Quebec, H4A 3J1, Canada
| |
Collapse
|
128
|
Dendritic cell extracellular vesicles. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 349:213-249. [DOI: 10.1016/bs.ircmb.2019.08.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
129
|
The Crosstalk between Cancer Stem Cells and Microenvironment Is Critical for Solid Tumor Progression: The Significant Contribution of Extracellular Vesicles. Stem Cells Int 2018; 2018:6392198. [PMID: 30532788 PMCID: PMC6247433 DOI: 10.1155/2018/6392198] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/02/2018] [Accepted: 10/11/2018] [Indexed: 12/12/2022] Open
Abstract
Several evidences nowadays demonstrated the critical role of the microenvironment in regulating cancer stem cells and their involvement in tumor progression. Extracellular vesicles (EVs) are considered as one of the most effective vehicles of information among cells. Accordingly, a number of studies led to the recognition of stem cell-associated EVs as new complexes able to contribute to cell fate determination of either normal or tumor cells. In this review, we aim to highlight an existing bidirectional role of EV-mediated communication—from cancer stem cells to microenvironment and also from microenvironment to cancer stem cells—in the most widespread solid cancers as prostate, breast, lung, and colon tumors.
Collapse
|
130
|
Reis-Sobreiro M, Chen JF, Novitskaya T, You S, Morley S, Steadman K, Gill NK, Eskaros A, Rotinen M, Chu CY, Chung LWK, Tanaka H, Yang W, Knudsen BS, Tseng HR, Rowat AC, Posadas EM, Zijlstra A, Di Vizio D, Freeman MR. Emerin Deregulation Links Nuclear Shape Instability to Metastatic Potential. Cancer Res 2018; 78:6086-6097. [PMID: 30154147 DOI: 10.1158/0008-5472.can-18-0608] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 06/13/2018] [Accepted: 08/22/2018] [Indexed: 01/21/2023]
Abstract
Abnormalities in nuclear shape are a well-known feature of cancer, but their contribution to malignant progression remains poorly understood. Here, we show that depletion of the cytoskeletal regulator, Diaphanous-related formin 3 (DIAPH3), or the nuclear membrane-associated proteins, lamin A/C, in prostate and breast cancer cells, induces nuclear shape instability, with a corresponding gain in malignant properties, including secretion of extracellular vesicles that contain genomic material. This transformation is characterized by a reduction and/or mislocalization of the inner nuclear membrane protein, emerin. Consistent with this, depletion of emerin evokes nuclear shape instability and promotes metastasis. By visualizing emerin localization, evidence for nuclear shape instability was observed in cultured tumor cells, in experimental models of prostate cancer, in human prostate cancer tissues, and in circulating tumor cells from patients with metastatic disease. Quantitation of emerin mislocalization discriminated cancer from benign tissue and correlated with disease progression in a prostate cancer cohort. Taken together, these results identify emerin as a mediator of nuclear shape stability in cancer and show that destabilization of emerin can promote metastasis.Significance: This study identifies a novel mechanism integrating the control of nuclear structure with the metastatic phenotype, and our inclusion of two types of human specimens (cancer tissues and circulating tumor cells) demonstrates direct relevance to human cancer.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/21/6086/F1.large.jpg Cancer Res; 78(21); 6086-97. ©2018 AACR.
Collapse
Affiliation(s)
- Mariana Reis-Sobreiro
- Division of Cancer Biology and Therapeutics, Department of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jie-Fu Chen
- Division of Cancer Biology and Therapeutics, Department of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Tatiana Novitskaya
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee
| | - Sungyong You
- Division of Cancer Biology and Therapeutics, Department of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Samantha Morley
- Division of Cancer Biology and Therapeutics, Department of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Kenneth Steadman
- Division of Cancer Biology and Therapeutics, Department of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Navjot Kaur Gill
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California
| | - Adel Eskaros
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee
| | - Mirja Rotinen
- Division of Cancer Biology and Therapeutics, Department of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Chia-Yi Chu
- Urologic Oncology Program/Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Center Institute, Cedars-Sinai Medical Center, Los Angeles, California.,Division of Hematology/Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Leland W K Chung
- Urologic Oncology Program/Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Center Institute, Cedars-Sinai Medical Center, Los Angeles, California.,Division of Hematology/Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Hisashi Tanaka
- Division of Cancer Biology and Therapeutics, Department of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Wei Yang
- Division of Cancer Biology and Therapeutics, Department of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Beatrice S Knudsen
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California
| | - Amy C Rowat
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California
| | - Edwin M Posadas
- Urologic Oncology Program/Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Center Institute, Cedars-Sinai Medical Center, Los Angeles, California.,Division of Hematology/Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Andries Zijlstra
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee.,Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Dolores Di Vizio
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Michael R Freeman
- Division of Cancer Biology and Therapeutics, Department of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California.
| |
Collapse
|
131
|
Vagner T, Spinelli C, Minciacchi VR, Balaj L, Zandian M, Conley A, Zijlstra A, Freeman MR, Demichelis F, De S, Posadas EM, Tanaka H, Di Vizio D. Large extracellular vesicles carry most of the tumour DNA circulating in prostate cancer patient plasma. J Extracell Vesicles 2018; 7:1505403. [PMID: 30108686 PMCID: PMC6084494 DOI: 10.1080/20013078.2018.1505403] [Citation(s) in RCA: 245] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/28/2018] [Accepted: 07/24/2018] [Indexed: 12/11/2022] Open
Abstract
Cancer-derived extracellular vesicles (EVs) are membrane-enclosed structures of highly variable size. EVs contain a myriad of substances (proteins, lipid, RNA, DNA) that provide a reservoir of circulating molecules, thus offering a good source of biomarkers. We demonstrate here that large EVs (L-EV) (large oncosomes) isolated from prostate cancer (PCa) cells and patient plasma are an EV population that is enriched in chromosomal DNA, including large fragments up to 2 million base pair long. While L-EVs and small EVs (S-EV) (exosomes) isolated from the same cells contained similar amounts of protein, the DNA was more abundant in L-EV, despite S-EVs being more numerous. Consistent with in vitro observations, the abundance of DNA in L-EV obtained from PCa patient plasma was variable but frequently high. Conversely, negligible amounts of DNA were present in the S-EVs from the same patients. Controlled experimental conditions, with spike-ins of L-EVs and S-EVs from cancer cells in human plasma from healthy subjects, showed that circulating DNA is almost exclusively enclosed in L-EVs. Whole genome sequencing revealed that the DNA in L-EVs reflects genetic aberrations of the cell of origin, including copy number variations of genes frequently altered in metastatic PCa (i.e. MYC, AKT1, PTK2, KLF10 and PTEN). These results demonstrate that L-EV-derived DNA reflects the genomic make-up of the tumour of origin. They also support the conclusion that L-EVs are the fraction of plasma EVs with DNA content that should be interrogated for tumour-derived genomic alterations.
Collapse
Affiliation(s)
- Tatyana Vagner
- Department of Biomedical Sciences, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Samuel Oschin Comprehensive Cancer Institute, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Surgery, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Cristiana Spinelli
- Department of Biomedical Sciences, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Samuel Oschin Comprehensive Cancer Institute, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Surgery, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pediatrics, McGill University, The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Valentina R Minciacchi
- Department of Biomedical Sciences, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Samuel Oschin Comprehensive Cancer Institute, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Surgery, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Leonora Balaj
- Department of Neurology, Program in Neuroscience, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mandana Zandian
- Department of Biomedical Sciences, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Samuel Oschin Comprehensive Cancer Institute, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Surgery, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Andrew Conley
- Department of Pathology and Laboratory Medicine, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Andries Zijlstra
- Dep nartment of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael R Freeman
- Department of Biomedical Sciences, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Samuel Oschin Comprehensive Cancer Institute, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Surgery, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Medicine, University of California, Los Angeles, CA, USA
| | | | - Subhajyoti De
- Department of Pathology and Laboratory Medicine, Rutgers Cancer Institute, Rutgers the State University of New Jersey, New Brunswick, NJ, USA
| | - Edwin M Posadas
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hisashi Tanaka
- Department of Biomedical Sciences, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Samuel Oschin Comprehensive Cancer Institute, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Surgery, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dolores Di Vizio
- Department of Biomedical Sciences, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Samuel Oschin Comprehensive Cancer Institute, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Surgery, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
132
|
Momen-Heravi F, Getting SJ, Moschos SA. Extracellular vesicles and their nucleic acids for biomarker discovery. Pharmacol Ther 2018; 192:170-187. [PMID: 30081050 DOI: 10.1016/j.pharmthera.2018.08.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Extracellular vesicles (EVs) are a heterogenous population of vesicles originate from cells. EVs are found in different biofluids and carry different macromolecules, including proteins, lipids, and nucleic acids, providing a snap shot of the parental cells at the time of release. EVs have the ability to transfer molecular cargoes to other cells and can initiate different physiological and pathological processes. Mounting lines of evidence demonstrated that EVs' cargo and machinery is affected in disease states, positioning EVs as potential sources for the discovery of novel biomarkers. In this review, we demonstrate a conceptual overview of the EV field with particular focus on their nucleic acid cargoes. Current knowledge of EV subtypes, nucleic acid cargo and pathophysiological roles are outlined, with emphasis placed on advantages against competing analytes. We review the utility of EVs and their nucleic acid cargoes as biomarkers and critically assess the newly available advances in the field of EV biomarkers and high throughput technologies. Challenges to achieving the diagnostic potential of EVs, including sample handling, EV isolation, methodological considerations, and bioassay reproducibility are discussed. Future implementation of 'omics-based technologies and integration of systems biology approaches for the development of EV-based biomarkers and personalized medicine are also considered.
Collapse
Affiliation(s)
- Fatemeh Momen-Heravi
- Division of Periodontics, Section of Oral and Diagnostic Sciences, Columbia University, College of Dental Medicine, New York, NY, USA; Department of Biomedical Sciences, University of Westminster, London, UK.
| | - Stephen J Getting
- Department of Biomedical Sciences, University of Westminster, London, UK; Department of Life Sciences, University of Westminster, London, UK
| | - Sterghios Athanasios Moschos
- Department of Biomedical Sciences, University of Westminster, London, UK; Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle, UK
| |
Collapse
|
133
|
Jabalee J, Towle R, Garnis C. The Role of Extracellular Vesicles in Cancer: Cargo, Function, and Therapeutic Implications. Cells 2018; 7:cells7080093. [PMID: 30071693 PMCID: PMC6115997 DOI: 10.3390/cells7080093] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 07/27/2018] [Accepted: 07/29/2018] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous collection of membrane-bound structures that play key roles in intercellular communication. EVs are potent regulators of tumorigenesis and function largely via the shuttling of cargo molecules (RNA, DNA, protein, etc.) among cancer cells and the cells of the tumor stroma. EV-based crosstalk can promote proliferation, shape the tumor microenvironment, enhance metastasis, and allow tumor cells to evade immune destruction. In many cases these functions have been linked to the presence of specific cargo molecules. Herein we will review various types of EV cargo molecule and their functional impacts in the context of oncology.
Collapse
Affiliation(s)
- James Jabalee
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver V5Z 1L3, BC, Canada.
| | - Rebecca Towle
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver V5Z 1L3, BC, Canada.
| | - Cathie Garnis
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver V5Z 1L3, BC, Canada.
- Division of Otolaryngology, Department of Surgery, University of British Columbia, Vancouver V6T 1Z4, BC, Canada.
| |
Collapse
|
134
|
Karimi N, Cvjetkovic A, Jang SC, Crescitelli R, Hosseinpour Feizi MA, Nieuwland R, Lötvall J, Lässer C. Detailed analysis of the plasma extracellular vesicle proteome after separation from lipoproteins. Cell Mol Life Sci 2018; 75:2873-2886. [PMID: 29441425 PMCID: PMC6021463 DOI: 10.1007/s00018-018-2773-4] [Citation(s) in RCA: 337] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/03/2018] [Accepted: 02/01/2018] [Indexed: 12/15/2022]
Abstract
The isolation of extracellular vesicles (EVs) from blood is of great importance to understand the biological role of circulating EVs and to develop EVs as biomarkers of disease. Due to the concurrent presence of lipoprotein particles, however, blood is one of the most difficult body fluids to isolate EVs from. The aim of this study was to develop a robust method to isolate and characterise EVs from blood with minimal contamination by plasma proteins and lipoprotein particles. Plasma and serum were collected from healthy subjects, and EVs were isolated by size-exclusion chromatography (SEC), with most particles being present in fractions 8-12, while the bulk of the plasma proteins was present in fractions 11-28. Vesicle markers peaked in fractions 7-11; however, the same fractions also contained lipoprotein particles. The purity of EVs was improved by combining a density cushion with SEC to further separate lipoprotein particles from the vesicles, which reduced the contamination of lipoprotein particles by 100-fold. Using this novel isolation procedure, a total of 1187 proteins were identified in plasma EVs by mass spectrometry, of which several proteins are known as EV-associated proteins but have hitherto not been identified in the previous proteomic studies of plasma EVs. This study shows that SEC alone is unable to completely separate plasma EVs from lipoprotein particles. However, combining SEC with a density cushion significantly improved the separation of EVs from lipoproteins and allowed for a detailed analysis of the proteome of plasma EVs, thus making blood a viable source for EV biomarker discovery.
Collapse
Affiliation(s)
- Nasibeh Karimi
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Aleksander Cvjetkovic
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Su Chul Jang
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Codiak BioSciences, Cambridge, MA, 02139, USA
| | - Rossella Crescitelli
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Department of Clinical Chemistry, and Vesicle Observation Centre, Academic Medical Centre of the University of Amsterdam, Amsterdam, The Netherlands
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Lässer
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
135
|
Pös O, Biró O, Szemes T, Nagy B. Circulating cell-free nucleic acids: characteristics and applications. Eur J Hum Genet 2018; 26:937-945. [PMID: 29681621 PMCID: PMC6018748 DOI: 10.1038/s41431-018-0132-4] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/07/2018] [Accepted: 02/23/2018] [Indexed: 12/11/2022] Open
Abstract
Liquid biopsy is becoming a very popular sample obtaining procedure, replacing the invasive sampling methods for the diagnostic protocols. The advantages of this method include the possibility to isolate cell-free nucleic acids (cfNAs) for diagnostic or screening purposes. A comprehensive review combining all current and perspective applications of cell-free nucleic acids is missing. Published articles are dealing with one type of cfNAs, or discuss them from the perspective of single disorder. We collected here all known types of cfNAs which are known to be present in biological fluids and could be involved in further studies to find out the exact biological role of them in normal physiological and pathological conditions. Beyond doubt, cfNAs will have a tremendous effect in future screening, diagnosis, prognosis, follow-up and treatment of cardiovascular diseases, cancer, diabetes and other diseases.
Collapse
Affiliation(s)
- Ondrej Pös
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| | - Orsolya Biró
- Genetic Laboratory, 1st Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| | - Tomas Szemes
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| | - Bálint Nagy
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
136
|
Giulietti M, Santoni M, Cimadamore A, Carrozza F, Piva F, Cheng L, Lopez-Beltran A, Scarpelli M, Battelli N, Montironi R. Exploring Small Extracellular Vesicles for Precision Medicine in Prostate Cancer. Front Oncol 2018; 8:221. [PMID: 29951374 PMCID: PMC6008382 DOI: 10.3389/fonc.2018.00221] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/29/2018] [Indexed: 12/21/2022] Open
Abstract
Tumor microenvironment constitutes a complex network in which tumor cells communicate among them and with stromal and immune cells. It has been shown that cancer cells are able to exchange genetic materials through small extracellular vesicles (EVs), a heterogeneous group of vesicles with different size and shape, cargo content, and function. The importance to investigate populations of circulating EVs would be of great importance as prostate cancer (PCa) biomarkers. In several neoplasms as well as in PCa, nanometer-sized EVs of endosomal origin are implicated in supporting tumor growth and metastatic spread by both altering local stroma cells and creating a protumor environment that favors the formation of pre-metastatic niches. Several techniques are applicable for the isolation and analysis of PCa-derived small EVs and are illustrated in this article. Due to the high sensitivity and specificity of these techniques, small EVs have become ideal candidates for early diagnosis. Moreover, we discuss the role of small EVs during PCa carcinogenesis, as well as in modulating the development of drug resistance to hormonal therapy and chemotherapy, thus underlining the potential of EV-tailored strategies in PCa patients.
Collapse
Affiliation(s)
- Matteo Giulietti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | | | - Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, Ancona, Italy
| | | | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | - Marina Scarpelli
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, Ancona, Italy
| | | | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, Ancona, Italy
| |
Collapse
|
137
|
Vlaeminck-Guillem V. Extracellular Vesicles in Prostate Cancer Carcinogenesis, Diagnosis, and Management. Front Oncol 2018; 8:222. [PMID: 29951375 PMCID: PMC6008571 DOI: 10.3389/fonc.2018.00222] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 05/29/2018] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs), especially exosomes, are now well recognized as major ways by which cancer cells interact with each other and stromal cells. The meaningful messages transmitted by the EVs are carried by all components of the EVs, i.e., the membrane lipids and the cargo (DNAs, RNAs, microRNAs, long non-coding RNAs, proteins). They are clearly part of the armed arsenal by which cancer cells obtain and share more and more advantages to grow and conquer new spaces. Identification of these messages offers a significant opportunity to better understand how a cancer occurs and then develops both locally and distantly. But it also provides a powerful means by which cancer progression can be detected and monitored. In the last few years, significant research efforts have been made to precisely identify how the EV trafficking is modified in cancer cells as compared to normal cells and how this trafficking is altered during cancer progression. Prostate cancer has not escaped this trend. The aim of this review is to describe the results obtained when assessing the meaningful content of prostate cancer- and stromal-derived EVs in terms of a better comprehension of the cellular and molecular mechanisms underlying prostate cancer occurrence and development. This review also deals with the use of EVs as powerful tools to diagnose non-indolent prostate cancer as early as possible and to accurately define, in a personalized approach, its present and potential aggressiveness, its response to treatment (androgen deprivation, chemotherapy, radiation, surgery), and the overall patients’ prognosis.
Collapse
Affiliation(s)
- Virginie Vlaeminck-Guillem
- Medical Unit of Molecular Oncology and Transfer, Department of Biochemistry and Molecular Biology, Centre Hospitalier Lyon-Sud, Hospices Civils of Lyon, Pierre-Bénite, France.,Cancer Research Centre of Lyon, U1052 INSERM, CNRS 5286, Claude Bernard University Lyon 1, Léon Bérard Centre, Lyon, France
| |
Collapse
|
138
|
Simon C, Greening DW, Bolumar D, Balaguer N, Salamonsen LA, Vilella F. Extracellular Vesicles in Human Reproduction in Health and Disease. Endocr Rev 2018; 39:292-332. [PMID: 29390102 DOI: 10.1210/er.2017-00229] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 01/25/2018] [Indexed: 02/07/2023]
Abstract
Extensive evidence suggests that the release of membrane-enclosed compartments, more commonly known as extracellular vesicles (EVs), is a potent newly identified mechanism of cell-to-cell communication both in normal physiology and in pathological conditions. This review presents evidence about the formation and release of different EVs, their definitive markers and cargo content in reproductive physiological processes, and their capacity to convey information between cells through the transfer of functional protein and genetic information to alter phenotype and function of recipient cells associated with reproductive biology. In the male reproductive tract, epididymosomes and prostasomes participate in regulating sperm motility activation, capacitation, and acrosome reaction. In the female reproductive tract, follicular fluid, oviduct/tube, and uterine cavity EVs are considered as vehicles to carry information during oocyte maturation, fertilization, and embryo-maternal crosstalk. EVs via their cargo might be also involved in the triggering, maintenance, and progression of reproductive- and obstetric-related pathologies such as endometriosis, polycystic ovarian syndrome, preeclampsia, gestational diabetes, and erectile dysfunction. In this review, we provide current knowledge on the present and future use of EVs not only as biomarkers, but also as therapeutic targeting agents, mainly as vectors for drug or compound delivery into target cells and tissues.
Collapse
Affiliation(s)
- Carlos Simon
- Igenomix Foundation, Valencia, Spain.,Instituto de Investigación Sanitaria Hospital Clínico (INCLIVA), Valencia, Spain.,Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, Valencia University, Valencia, Spain.,Department of Obstetrics and Gynecology, Stanford University, Palo Alto, California
| | - David W Greening
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - David Bolumar
- Igenomix Foundation, Valencia, Spain.,Instituto de Investigación Sanitaria Hospital Clínico (INCLIVA), Valencia, Spain
| | - Nuria Balaguer
- Igenomix Foundation, Valencia, Spain.,Instituto de Investigación Sanitaria Hospital Clínico (INCLIVA), Valencia, Spain
| | - Lois A Salamonsen
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Felipe Vilella
- Igenomix Foundation, Valencia, Spain.,Instituto de Investigación Sanitaria Hospital Clínico (INCLIVA), Valencia, Spain.,Department of Obstetrics and Gynecology, Stanford University, Palo Alto, California
| |
Collapse
|
139
|
Konečná B, Lauková L, Vlková B. Immune activation by nucleic acids: A role in pregnancy complications. Scand J Immunol 2018; 87:e12651. [PMID: 29479732 DOI: 10.1111/sji.12651] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 02/15/2018] [Indexed: 12/25/2022]
Abstract
Cell-free self-DNA or RNA may induce an immune response by activating specific sensing receptors. During pregnancy, placental nucleic acids present in the maternal circulation further activate these receptors due to the presence of unmethylated CpG islands. A higher concentration of cell-free foetal DNA is associated with pregnancy complications and a higher risk for foetal rejection. Cell-free foetal DNA originates from placental trophoblasts. It appears in different forms: free, bound to histones in nucleosomes, in neutrophil extracellular traps (NETs) and in extracellular vesicles (EVs). In several pregnancy complications, cell-free foetal DNA triggers the production of proinflammatory cytokines, and this production results in a cellular and humoral immune response. This review discusses preeclampsia, systemic lupus erythematosus, foetal growth restriction, gestational diabetes, rheumatoid arthritis and obesity in pregnancy from an immunological point of view and closely examines the different pathways that result in maternal inflammation. Understanding the role of cell-free nucleic acids, as well as the biogenesis of NETs and EVs, will help us to specify their functions or targets, which seem to be important in pregnancy complications. It is still not clear whether higher concentrations of cell-free nucleic acids in the maternal circulation are the cause or consequence of various complications. Therefore, further clinical studies and, even more importantly, animal experiments that focus on the involved immunological pathways are needed.
Collapse
Affiliation(s)
- B Konečná
- Faculty of Medicine, Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia
| | - L Lauková
- Faculty of Medicine, Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia
| | - B Vlková
- Faculty of Medicine, Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
140
|
Garcia-Romero N, Esteban-Rubio S, Rackov G, Carrión-Navarro J, Belda-Iniesta C, Ayuso-Sacido A. Extracellular vesicles compartment in liquid biopsies: Clinical application. Mol Aspects Med 2018; 60:27-37. [DOI: 10.1016/j.mam.2017.11.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/08/2017] [Accepted: 11/14/2017] [Indexed: 02/07/2023]
|
141
|
Nogués L, Benito-Martin A, Hergueta-Redondo M, Peinado H. The influence of tumour-derived extracellular vesicles on local and distal metastatic dissemination. Mol Aspects Med 2018; 60:15-26. [PMID: 29196097 PMCID: PMC5856602 DOI: 10.1016/j.mam.2017.11.012] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/18/2017] [Accepted: 11/27/2017] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) are key mediators of intercellular communication that have been ignored for decades. Tumour cells benefit from the secretion of vesicles as they can influence the behaviour of neighbouring tumour cells within the tumour microenvironment. Several studies have shown that extracellular vesicles play an active role in pre-metastatic niche formation and importantly, they are involved in the metastatic organotropism of different tumour types. Tumour-derived EVs carry and transfer molecules to recipient cells, modifying their behaviour through a process defined as "EV-driven education". EVs favour metastasis to sentinel lymph nodes and distal organs by reinforcing angiogenesis, inflammation and lymphangiogenesis. Hence, in this review we will summarize the main mechanisms by which tumour-derived EVs regulate lymph node and distal organ metastasis. Moreover, since some cancers metastasize through the lymphatic system, we will discuss recent discoveries about the presence and function of tumour EVs in the lymph. Finally, we will address the potential value of tumour EVs as prognostic biomarkers in liquid biopsies, specially blood and lymphatic fluid, and the use of these tools as early detectors of metastases.
Collapse
Affiliation(s)
- Laura Nogués
- Children's Cancer and Blood Foundation Laboratories, Department of Pediatrics, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY 10021, USA
| | - Alberto Benito-Martin
- Children's Cancer and Blood Foundation Laboratories, Department of Pediatrics, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY 10021, USA
| | - Marta Hergueta-Redondo
- Microenvironment and Metastasis Group, Department of Molecular Oncology, Spanish National Cancer Research Center (CNIO), Madrid 28029, Spain
| | - Héctor Peinado
- Children's Cancer and Blood Foundation Laboratories, Department of Pediatrics, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY 10021, USA; Microenvironment and Metastasis Group, Department of Molecular Oncology, Spanish National Cancer Research Center (CNIO), Madrid 28029, Spain.
| |
Collapse
|
142
|
García-Romero N, Carrión-Navarro J, Esteban-Rubio S, Lázaro-Ibáñez E, Peris-Celda M, Alonso MM, Guzmán-De-Villoria J, Fernández-Carballal C, de Mendivil AO, García-Duque S, Escobedo-Lucea C, Prat-Acín R, Belda-Iniesta C, Ayuso-Sacido A. DNA sequences within glioma-derived extracellular vesicles can cross the intact blood-brain barrier and be detected in peripheral blood of patients. Oncotarget 2018; 8:1416-1428. [PMID: 27902458 PMCID: PMC5352065 DOI: 10.18632/oncotarget.13635] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/07/2016] [Indexed: 01/01/2023] Open
Abstract
Tumor-cell-secreted extracellular vesicles (EVs) can cross the disrupted blood-brain barrier (BBB) into the bloodstream. However, in certain gliomas, the BBB remains intact, which might limit EVs release. To evaluate the ability of tumor-derived EVs to cross the BBB, we used an orthotopic xenotransplant mouse model of human glioma-cancer stem cells featuring an intact BBB. We demonstrated that all types of tumor cells-derived EVs−apoptotic bodies, shedding microvesicles and exosomes−cross the intact BBB and can be detected in the peripheral blood, which provides a minimally invasive method for their detection compared to liquid biopsies obtained from cerebrospinal fluid (CSF). Furthermore, these EVs can be readily distinguished from total murine EVs, since they carry human-specific DNA sequences relevant for GBM biology. In a small cohort of glioma patients, we finally demonstrated that peripheral blood EVs cargo can be successfully used to detect the presence of IDH1G395A, an essential biomarker in the current management of human glioma
Collapse
Affiliation(s)
- Noemí García-Romero
- Fundación de Investigación HM Hospitales, HM Hospitales, Madrid, Spain.,IMDEA Nanoscience, Madrid, Spain
| | - Josefa Carrión-Navarro
- Fundación de Investigación HM Hospitales, HM Hospitales, Madrid, Spain.,Facultad de Medicina (IMMA), Universidad San Pablo-CEU, Madrid, Spain
| | - Susana Esteban-Rubio
- Fundación de Investigación HM Hospitales, HM Hospitales, Madrid, Spain.,Facultad de Medicina (IMMA), Universidad San Pablo-CEU, Madrid, Spain
| | - Elisa Lázaro-Ibáñez
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - María Peris-Celda
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Juan Guzmán-De-Villoria
- Servicio de Radiodiagnóstico, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | | | | - Sara García-Duque
- Fundación de Investigación HM Hospitales, HM Hospitales, Madrid, Spain
| | - Carmen Escobedo-Lucea
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Ricardo Prat-Acín
- Departamento de Neurocirugía, Hospital Universitario la Fe, Valencia, Spain
| | - Cristóbal Belda-Iniesta
- Fundación de Investigación HM Hospitales, HM Hospitales, Madrid, Spain.,Facultad de Medicina (IMMA), Universidad San Pablo-CEU, Madrid, Spain
| | - Angel Ayuso-Sacido
- Fundación de Investigación HM Hospitales, HM Hospitales, Madrid, Spain.,IMDEA Nanoscience, Madrid, Spain.,Facultad de Medicina (IMMA), Universidad San Pablo-CEU, Madrid, Spain
| |
Collapse
|
143
|
Osteikoetxea X, Benke M, Rodriguez M, Pálóczi K, Sódar BW, Szvicsek Z, Szabó-Taylor K, Vukman KV, Kittel Á, Wiener Z, Vékey K, Harsányi L, Szűcs Á, Turiák L, Buzás EI. Detection and proteomic characterization of extracellular vesicles in human pancreatic juice. Biochem Biophys Res Commun 2018; 499:37-43. [PMID: 29550476 DOI: 10.1016/j.bbrc.2018.03.107] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 03/13/2018] [Indexed: 02/07/2023]
Abstract
AIMS The prognosis of patients with pancreatic cancer has remained virtually unchanged with a high mortality rate compared to other types of cancers. An earlier detection would provide a time window of opportunity for treatment and prevention of deaths. In the present study we investigated extracellular vesicle (EV)-associated potential biomarkers for pancreatic cancer by directly assessing EV size-based subpopulations in pancreatic juice samples of patients with chronic pancreatitis or pancreatic cancer. In addition, we also studied blood plasma and pancreatic cancer cell line-derived EVs. METHODS Comparative proteomic analysis was performed of 102 EV preparations from human pancreatic juices, blood, and pancreatic cancer cell lines Capan-1 and MIA PaCa-2. EV preparations were also characterized by electron microscopy, tunable resistive pulse sensing, and flow cytometry. RESULTS Here we describe the presence of EVs in human pancreatic juice samples. Pancreatic juice EV-associated proteins that we identified as possible candidate markers for pancreatic cancer included mucins, such as MUC1, MUC4, MUC5AC, MUC6 and MUC16, CFTR, and MDR1 proteins. These candidate biomarkers could also be detected by flow cytometry in EVs found in pancreatic juice and those secreted by pancreatic cancer cell lines. CONCLUSIONS Together our data show that detection and characterization of EVs directly in pancreatic juice is feasible and may prove to be a valuable source of potential biomarkers of pancreatic cancer.
Collapse
Affiliation(s)
- Xabier Osteikoetxea
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary.
| | - Márton Benke
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary; 1st Department of Surgery, Semmelweis University, Budapest, Hungary
| | - Marta Rodriguez
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Krisztina Pálóczi
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Barbara W Sódar
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Zsuzsanna Szvicsek
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Katalin Szabó-Taylor
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Krisztina V Vukman
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Ágnes Kittel
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Zoltán Wiener
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Károly Vékey
- Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest Hungary
| | - László Harsányi
- 1st Department of Surgery, Semmelweis University, Budapest, Hungary
| | - Ákos Szűcs
- 1st Department of Surgery, Semmelweis University, Budapest, Hungary
| | - Lilla Turiák
- Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest Hungary
| | - Edit I Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary; MTA-SE Immune-Proteogenomics Extracellular Vesicles Research Group, Budapest, Hungary.
| |
Collapse
|
144
|
Yang MQ, Du Q, Goswami J, Varley PR, Chen B, Wang RH, Morelli AE, Stolz DB, Billiar TR, Li J, Geller DA. Interferon regulatory factor 1-Rab27a regulated extracellular vesicles promote liver ischemia/reperfusion injury. Hepatology 2018; 67:1056-1070. [PMID: 29059701 PMCID: PMC5826835 DOI: 10.1002/hep.29605] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 10/06/2017] [Accepted: 10/16/2017] [Indexed: 12/11/2022]
Abstract
The role and regulators of extracellular vesicle (EV) secretion in hepatic ischemia/reperfusion (IR) injury have not been defined. Rab27a is a guanosine triphosphatase known to control EV release. Interferon regulatory factor 1 (IRF-1) is a transcription factor that plays an important role in liver IR and regulates certain guanosine triphosphatases. However, the relationships among IRF-1, Rab27a, and EV secretion are largely unknown. Here, we show induction of IRF-1 and Rab27a both in vitro in hypoxic hepatocytes and in vivo in warm IR and orthotopic liver transplantation livers. Interferon γ stimulation, IRF-1 transduction, or IR promoted Rab27a expression and EV secretion. Meanwhile, silencing of IRF-1 decreased Rab27a expression and EV secretion. Rab27a silencing decreased EV secretion and liver IR injury. Ten putative IRF-1 binding motifs in the 1,692-bp Rab27a promoter region were identified. Chromatin immunoprecipitation and electrophoretic mobility shift assay verified five functional IRF-1 binding motifs, which were confirmed by a Rab27a promoter luciferase assay. IR-induced EVs contained higher oxidized phospholipids (OxPL). OxPLs on the EV surface activated neutrophils through the toll-like receptor 4 pathway. OxPL-neutralizing E06 antibody blocked the effect of EVs and decreased liver IR injury. CONCLUSION These findings provide a novel mechanism by which IRF-1 regulates Rab27a transcription and EV secretion, leading to OxPL activation of neutrophils and subsequent hepatic IR injury. (Hepatology 2018;67:1056-1070).
Collapse
Affiliation(s)
- Mu-qing Yang
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, 200 Lothrop Street, W1507, Pittsburgh, PA 15261
- Department of Surgery, Shanghai Tenth People’s Hospital, Tenth People’s Hospital of Tongji University, 301 Middle Yanchang Road, Shanghai 200072, People’s Republic of China
| | - Qiang Du
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, 200 Lothrop Street, W1507, Pittsburgh, PA 15261
| | - Julie Goswami
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, 200 Lothrop Street, W1507, Pittsburgh, PA 15261
| | - Patrick R. Varley
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, 200 Lothrop Street, W1507, Pittsburgh, PA 15261
| | - Bin Chen
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, 200 Lothrop Street, W1507, Pittsburgh, PA 15261
| | - Rong-hua Wang
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, 200 Lothrop Street, W1507, Pittsburgh, PA 15261
| | - Adrian E. Morelli
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, 200 Lothrop Street, W1507, Pittsburgh, PA 15261
| | - Donna B. Stolz
- Center of Biological Imaging, University of Pittsburgh, 3500 Terrace Street, BST S224, Pittsburgh, PA 15261
| | - Timothy R. Billiar
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, 200 Lothrop Street, W1507, Pittsburgh, PA 15261
| | - Jiyu Li
- Department of Surgery, Shanghai Tenth People’s Hospital, Tenth People’s Hospital of Tongji University, 301 Middle Yanchang Road, Shanghai 200072, People’s Republic of China
| | - David A. Geller
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, 200 Lothrop Street, W1507, Pittsburgh, PA 15261
| |
Collapse
|
145
|
Lässer C, Jang SC, Lötvall J. Subpopulations of extracellular vesicles and their therapeutic potential. Mol Aspects Med 2018; 60:1-14. [PMID: 29432782 DOI: 10.1016/j.mam.2018.02.002] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 01/17/2018] [Accepted: 02/06/2018] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EVs), such as exosomes and microvesicles, have over the last 10-15 years been recognized to convey key messages in the molecular communication between cells. Indeed, EVs have the capacity to shuttle proteins, lipids, and nucleotides such as RNA between cells, leading to an array of functional changes in the recipient cells. Importantly, the EV secretome changes significantly in diseased cells and under conditions of cellular stress. More recently, it has become evident that the EV secretome is exceptionally diverse, with many different types of EVs being released by a single cell type, and these EVs can be described in terms of differences in density, molecular cargos, and morphology. This review will discuss the diversity of EVs, will introduce some suggestions for how to categorize them, and will propose how EVs and their subpopulations might be used for very different therapeutic purposes.
Collapse
Affiliation(s)
- Cecilia Lässer
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Su Chul Jang
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Sweden; Codiak BioSciences, Cambridge, MA 02139, USA
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Sweden.
| |
Collapse
|
146
|
Conigliaro A, Fontana S, Raimondo S, Alessandro R. Exosomes: Nanocarriers of Biological Messages. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 998:23-43. [PMID: 28936730 DOI: 10.1007/978-981-10-4397-0_2] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cell-cell communication is crucial to maintain homeostasis in multicellular organism. Cells communicate each other by direct contact or by releasing factors that, soluble or packaged in membrane vesicles, can reach different regions of the organism. To date numerous studies highlighted the existence of several types of extracellular vesicles that, differing for dimension, origin and contents, play a role in physiological and/or pathological processes. Among extracellular vesicles, exosomes are emerging as efficient players to modulate target cells phenotype and as new non-invasive diagnostic and prognostic tools in multiple diseases. They, in fact, strictly reflect the type and functional status of the producing cells and are able to deliver their contents even over a long distance. The results accumulated in the last two decades and collected in this chapter, indicated that exosomes, can carry RNAs, microRNAs, long non-coding RNAs, DNA, lipids, metabolites and proteins; a deeper understanding of their contents is therefore needed to get the most from this incredible cell product.
Collapse
Affiliation(s)
- Alice Conigliaro
- Dipartimento di Biotecnologie Cellulari ed Ematologia, Sapienza University of Rome, Rome, 00185, Italy
- Dipartimento di Biopatologia e Biotecnologie Mediche, University of Palermo, Palermo, 90133, Italy
| | - Simona Fontana
- Dipartimento di Biopatologia e Biotecnologie Mediche, University of Palermo, Palermo, 90133, Italy
| | - Stefania Raimondo
- Dipartimento di Biopatologia e Biotecnologie Mediche, University of Palermo, Palermo, 90133, Italy
| | - Riccardo Alessandro
- Dipartimento di Biopatologia e Biotecnologie Mediche, University of Palermo, Palermo, 90133, Italy.
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council, Palermo, Italy.
| |
Collapse
|
147
|
Gustafson D, Veitch S, Fish JE. Extracellular Vesicles as Protagonists of Diabetic Cardiovascular Pathology. Front Cardiovasc Med 2017; 4:71. [PMID: 29209616 PMCID: PMC5701646 DOI: 10.3389/fcvm.2017.00071] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 10/26/2017] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) represent an emerging mechanism of cell–cell communication in the cardiovascular system. Recent data suggest that EVs are produced and taken up by multiple cardiovascular cell types, influencing target cells through signaling or transfer of cargo (including proteins, lipids, messenger RNA, and non-coding RNA). The concentration and contents of circulating EVs are altered in several diseases and represent explicit signatures of cellular activation, making them of particular interest as circulating biomarkers. EVs also actively contribute to the progression of various cardiovascular diseases, including diabetes-related vascular disease. Understanding the relationships between circulating EVs, diabetes, and cardiovascular disease is of importance as diabetic patients are at elevated risk for developing several debilitating cardiovascular pathologies, including diabetic cardiomyopathy (DCM), a disease that remains an enigma at the molecular level. Enhancing and exploiting our understanding of EV biology could facilitate the development of effective non-invasive diagnostics, prognostics, and therapeutics. This review will focus on EV biology in diabetic cardiovascular diseases, including atherosclerosis and DCM. We will review EV biogenesis and functional properties, as well as provide insight into their emerging role in cell–cell communication. Finally, we will address the utility of EVs as clinical biomarkers and outline their impact as a biomedical tool in the development of therapeutics.
Collapse
Affiliation(s)
- Dakota Gustafson
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Shawn Veitch
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jason E Fish
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.,Heart & Stroke Richard Lewar Center of Excellence in Cardiovascular Research, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
148
|
Angulski ABB, Capriglione LG, Batista M, Marcon BH, Senegaglia AC, Stimamiglio MA, Correa A. The Protein Content of Extracellular Vesicles Derived from Expanded Human Umbilical Cord Blood-Derived CD133 + and Human Bone Marrow-Derived Mesenchymal Stem Cells Partially Explains Why both Sources are Advantageous for Regenerative Medicine. Stem Cell Rev Rep 2017; 13:244-257. [PMID: 28054239 DOI: 10.1007/s12015-016-9715-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adult stem cells have beneficial effects when exposed to damaged tissue due, at least in part, to their paracrine activity, which includes soluble factors and extracellular vesicles (EVs). Given the multiplicity of signals carried by these vesicles through the horizontal transfer of functional molecules, human mesenchymal stem cell (hMSCs) and CD133+ cell-derived EVs have been tested in various disease models and shown to recover damaged tissues. In this study, we profiled the protein content of EVs derived from expanded human CD133+ cells and bone marrow-derived hMSCs with the intention of better understanding the functions performed by these vesicles/cells and delineating the most appropriate use of each EV in future therapeutic procedures. Using LC-MS/MS analysis, we identified 623 proteins for expanded CD133+-EVs and 797 proteins for hMSCs-EVs. Although the EVs from both origins were qualitatively similar, when protein abundance was considered, hMSCs-EVs and CD133+-EVs were different. Gene Ontology (GO) enrichment analysis in CD133+-EVs revealed proteins involved in a variety of angiogenesis-related functions as well proteins related to the cytoskeleton and highly implicated in cell motility and cellular activation. In contrast, when overrepresented proteins in hMSCs-EVs were analyzed, a GO cluster of immune response-related genes involved with immune response-regulating factors acting on phagocytosis and innate immunity was identified. Together our data demonstrate that from the point of view of protein content, expanded CD133+-EVs and hMSCs-EVs are in part similar but also sufficiently different to reflect the main beneficial paracrine effects widely reported in pre-clinical studies using expanded CD133+ cells and/or hBM-MSCs.
Collapse
Affiliation(s)
- Addeli B B Angulski
- Instituto Carlos Chagas, Fiocruz-Paraná, Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR, 81350-010, Brazil
| | - Luiz G Capriglione
- Núcleo de Tecnologia Celular, Pontifícia Universidade Católica do Paraná, Rua Imaculada Conceição, 1155, Curitiba, PR, 80215-901, Brazil
| | - Michel Batista
- Instituto Carlos Chagas, Fiocruz-Paraná, Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR, 81350-010, Brazil
| | - Bruna H Marcon
- Instituto Carlos Chagas, Fiocruz-Paraná, Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR, 81350-010, Brazil
| | - Alexandra C Senegaglia
- Núcleo de Tecnologia Celular, Pontifícia Universidade Católica do Paraná, Rua Imaculada Conceição, 1155, Curitiba, PR, 80215-901, Brazil
| | - Marco A Stimamiglio
- Instituto Carlos Chagas, Fiocruz-Paraná, Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR, 81350-010, Brazil.
| | - Alejandro Correa
- Instituto Carlos Chagas, Fiocruz-Paraná, Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR, 81350-010, Brazil.
| |
Collapse
|
149
|
Puhka M, Takatalo M, Nordberg ME, Valkonen S, Nandania J, Aatonen M, Yliperttula M, Laitinen S, Velagapudi V, Mirtti T, Kallioniemi O, Rannikko A, Siljander PRM, af Hällström TM. Metabolomic Profiling of Extracellular Vesicles and Alternative Normalization Methods Reveal Enriched Metabolites and Strategies to Study Prostate Cancer-Related Changes. Am J Cancer Res 2017; 7:3824-3841. [PMID: 29109780 PMCID: PMC5667407 DOI: 10.7150/thno.19890] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 07/06/2017] [Indexed: 12/11/2022] Open
Abstract
Body fluids are a rich source of extracellular vesicles (EVs), which carry cargo derived from the secreting cells. So far, biomarkers for pathological conditions have been mainly searched from their protein, (mi)RNA, DNA and lipid cargo. Here, we explored the small molecule metabolites from urinary and platelet EVs relative to their matched source samples. As a proof-of-concept study of intra-EV metabolites, we compared alternative normalization methods to profile urinary EVs from prostate cancer patients before and after prostatectomy and from healthy controls. Methods: We employed targeted ultra-performance liquid chromatography-tandem mass spectrometry to profile over 100 metabolites in the isolated EVs, original urine samples and platelets. We determined the enrichment of the metabolites in the EVs and analyzed their subcellular origin, pathways and relevant enzymes or transporters through data base searches. EV- and urine-derived factors and ratios between metabolites were tested for normalization of the metabolomics data. Results: Approximately 1 x 1010 EVs were sufficient for detection of metabolite profiles from EVs. The profiles of the urinary and platelet EVs overlapped with each other and with those of the source materials, but they also contained unique metabolites. The EVs enriched a selection of cytosolic metabolites including members from the nucleotide and spermidine pathways, which linked to a number of EV-resident enzymes or transporters. Analysis of the urinary EVs from the patients indicated that the levels of glucuronate, D-ribose 5-phosphate and isobutyryl-L-carnitine were 2-26-fold lower in all pre-prostatectomy samples compared to the healthy control and post-prostatectomy samples (p < 0.05). These changes were only detected from EVs by normalization to EV-derived factors or with metabolite ratios, and not from the original urine samples. Conclusions: Our results suggest that metabolite analysis of EVs from different samples is feasible using a high-throughput platform and relatively small amount of sample material. With the knowledge about the specific enrichment of metabolites and normalization methods, EV metabolomics could be used to gain novel biomarker data not revealed by the analysis of the original EV source materials.
Collapse
|
150
|
Coumans FAW, Brisson AR, Buzas EI, Dignat-George F, Drees EEE, El-Andaloussi S, Emanueli C, Gasecka A, Hendrix A, Hill AF, Lacroix R, Lee Y, van Leeuwen TG, Mackman N, Mäger I, Nolan JP, van der Pol E, Pegtel DM, Sahoo S, Siljander PRM, Sturk G, de Wever O, Nieuwland R. Methodological Guidelines to Study Extracellular Vesicles. Circ Res 2017; 120:1632-1648. [PMID: 28495994 DOI: 10.1161/circresaha.117.309417] [Citation(s) in RCA: 698] [Impact Index Per Article: 99.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Owing to the relationship between extracellular vesicles (EVs) and physiological and pathological conditions, the interest in EVs is exponentially growing. EVs hold high hopes for novel diagnostic and translational discoveries. This review provides an expert-based update of recent advances in the methods to study EVs and summarizes currently accepted considerations and recommendations from sample collection to isolation, detection, and characterization of EVs. Common misconceptions and methodological pitfalls are highlighted. Although EVs are found in all body fluids, in this review, we will focus on EVs from human blood, not only our most complex but also the most interesting body fluid for cardiovascular research.
Collapse
Affiliation(s)
- Frank A W Coumans
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Alain R Brisson
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Edit I Buzas
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Françoise Dignat-George
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Esther E E Drees
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Samir El-Andaloussi
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Costanza Emanueli
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Aleksandra Gasecka
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - An Hendrix
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Andrew F Hill
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Romaric Lacroix
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Yi Lee
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Ton G van Leeuwen
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Nigel Mackman
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Imre Mäger
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - John P Nolan
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Edwin van der Pol
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - D Michiel Pegtel
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Susmita Sahoo
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Pia R M Siljander
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Guus Sturk
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Olivier de Wever
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Rienk Nieuwland
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.).
| |
Collapse
|