1
|
Sözmen A, Arslan-Yildiz A. Utilizing Magnetic Levitation to Detect Lung Cancer-Associated Exosomes. ACS Sens 2024; 9:2043-2049. [PMID: 38520356 PMCID: PMC11059084 DOI: 10.1021/acssensors.4c00011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/18/2024] [Accepted: 03/11/2024] [Indexed: 03/25/2024]
Abstract
Extracellular vesicles, especially exosomes, have attracted attention in the last few decades as novel cancer biomarkers. Exosomal membrane proteins provide easy-to-reach targets and can be utilized as information sources of their parent cells. In this study, a MagLev-based, highly sensitive, and versatile biosensor platform for detecting minor differences in the density of suspended objects is proposed for exosome detection. The developed platform utilizes antibody-functionalized microspheres to capture exosomal membrane proteins (ExoMPs) EpCAM, CD81, and CD151 as markers for cancerous exosomes, exosomes, and non-small cell lung cancer (NSCLC)-derived exosomes, respectively. Initially, the platform was utilized for protein detection and quantification by targeting solubilized ExoMPs, and a dynamic range of 1-100 nM, with LoD values of 1.324, 0.638, and 0.722 nM for EpCAM, CD81, and CD151, were observed, respectively. Then, the sensor platform was tested using exosome isolates derived from NSCLC cell line A549 and MRC5 healthy lung fibroblast cell line. It was shown that the sensor platform is able to detect and differentiate exosomal biomarkers derived from cancerous and non-cancerous cell lines. Overall, this innovative, simple, and rapid method shows great potential for the early diagnosis of lung cancer through exosomal biomarker detection.
Collapse
Affiliation(s)
- Alper
Baran Sözmen
- Bioengineering Department, Izmir Institute of Technology, 35430 Izmir, Turkey
| | - Ahu Arslan-Yildiz
- Bioengineering Department, Izmir Institute of Technology, 35430 Izmir, Turkey
| |
Collapse
|
2
|
Yung C, Zhang Y, Kuhn M, Armstrong RJ, Olyaei A, Aloia M, Scottoline B, Andres SF. Neonatal enteroids absorb extracellular vesicles from human milk-fed infant digestive fluid. J Extracell Vesicles 2024; 13:e12422. [PMID: 38602306 PMCID: PMC11007820 DOI: 10.1002/jev2.12422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 02/20/2024] [Indexed: 04/12/2024] Open
Abstract
Human milk contains extracellular vesicles (HMEVs). Pre-clinical models suggest that HMEVs may enhance intestinal function and limit inflammation; however, it is unknown if HMEVs or their cargo survive neonatal human digestion. This limits the ability to leverage HMEV cargo as additives to infant nutrition or as therapeutics. This study aimed to develop an EV isolation pipeline from small volumes of human milk and neonatal intestinal contents after milk feeding (digesta) to address the hypothesis that HMEVs survive in vivo neonatal digestion to be taken up intestinal epithelial cells (IECs). Digesta was collected from nasoduodenal sampling tubes or ostomies. EVs were isolated from raw and pasteurized human milk and digesta by density-gradient ultracentrifugation following two-step skimming, acid precipitation of caseins, and multi-step filtration. EVs were validated by electron microscopy, western blotting, nanoparticle tracking analysis, resistive pulse sensing, and super-resolution microscopy. EV uptake was tested in human neonatal enteroids. HMEVs and digesta EVs (dEVs) show typical EV morphology and are enriched in CD81 and CD9, but depleted of β-casein and lactalbumin. HMEV and some dEV fractions contain mammary gland-derived protein BTN1A1. Neonatal human enteroids rapidly take up dEVs in part via clathrin-mediated endocytosis. Our data suggest that EVs can be isolated from digestive fluid and that these dEVs can be absorbed by IECs.
Collapse
Affiliation(s)
- Claire Yung
- Department of PediatricsPediatric GI Division, School of Medicine, Oregon Health and Science UniversityPortlandOregonUSA
| | - Yang Zhang
- Department of PediatricsPediatric GI Division, School of Medicine, Oregon Health and Science UniversityPortlandOregonUSA
| | - Madeline Kuhn
- Department of PediatricsPediatric GI Division, School of Medicine, Oregon Health and Science UniversityPortlandOregonUSA
| | - Randall J. Armstrong
- Knight Cancer InstituteOregon Health and Science UniversityPortlandOregonUSA
- Cancer Early Detection Advanced Research (CEDAR)Oregon Health and Science UniversityPortlandOregonUSA
| | - Amy Olyaei
- Division of Neonatology, Department of PediatricsOregon Health and Science UniversityPortlandOregonUSA
| | - Molly Aloia
- Division of Neonatology, Department of PediatricsOregon Health and Science UniversityPortlandOregonUSA
| | - Brian Scottoline
- Department of PediatricsPediatric GI Division, School of Medicine, Oregon Health and Science UniversityPortlandOregonUSA
- Division of Neonatology, Department of PediatricsOregon Health and Science UniversityPortlandOregonUSA
| | - Sarah F. Andres
- Department of PediatricsPediatric GI Division, School of Medicine, Oregon Health and Science UniversityPortlandOregonUSA
| |
Collapse
|
3
|
Mantel M, Durand T, Bessard A, Pernet S, Beaudeau J, Guimaraes-Laguna J, Maillard MB, Guédon E, Neunlist M, Le Loir Y, Jan G, Rolli-Derkinderen M. Propionibacterium freudenreichii CIRM-BIA 129 mitigates colitis through S layer protein B-dependent epithelial strengthening. Am J Physiol Gastrointest Liver Physiol 2024; 326:G163-G175. [PMID: 37988603 DOI: 10.1152/ajpgi.00198.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/10/2023] [Accepted: 11/20/2023] [Indexed: 11/23/2023]
Abstract
The growing incidence of human diseases involving inflammation and increased gut permeability makes the quest for protective functional foods more crucial than ever. Propionibacterium freudenreichii (P. freudenreichii) is a beneficial bacterium used in the dairy and probiotic industries. Selected strains exert anti-inflammatory effects, and the present work addresses whether the P. freudenreichii CIRM-BIA129, consumed daily in a preventive way, could protect mice from acute colitis induced by dextran sodium sulfate (DSS), and more precisely, whether it could protect from intestinal epithelial breakdown induced by inflammation. P. freudenreichii CIRM-BIA129 mitigated colitis severity and inhibited DSS-induced permeability. It limited crypt length reduction and promoted the expression of zonula occludens-1 (ZO-1), without reducing interleukin-1β mRNA (il-1β) expression. In vitro, P. freudenreichii CIRM-BIA129 prevented the disruption of a Caco-2 monolayer induced by proinflammatory cytokines. It increased transepithelial electrical resistance (TEER) and inhibited permeability induced by inflammation, along with an increased ZO-1 expression. Extracellular vesicles (EVs) from P. freudenreichii CIRM-BIA129, carrying the surface layer protein (SlpB), reproduced the protective effect of P. freudenreichii CIRM-BIA129. A mutant strain deleted for slpB (ΔslpB), or EVs from this mutant strain, had lost their protective effects and worsened both DSS-induced colitis and inflammation in vivo. These results shown that P. freudenreichii CIRM-BIA129 daily consumption has the potential to greatly alleviate colitis symptoms and, particularly, to counter intestinal epithelial permeability induced by inflammation by restoring ZO-1 expression through mechanisms involving S-layer protein B. They open new avenues for the use of probiotic dairy propionibacteria and/or postbiotic fractions thereof, in the context of gut permeability.NEW & NOTEWORTHY Propionibacterium freudenreichii reduces dextran sodium sulfate (DSS)-induced intestinal permeability in vivo. P. freudenreichii does not inhibit inflammation but damages linked to inflammation. P. freudenreichii inhibits intestinal epithelial breakdown through S-layer protein B. The protective effects of P. freudenreichii depend on S-layer protein B. Extracellular vesicles from P. freudenreichii CB 129 mimic the protective effect of the probiotic.
Collapse
Affiliation(s)
- Marine Mantel
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Tony Durand
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| | - Anne Bessard
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| | - Ségolène Pernet
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| | - Julie Beaudeau
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
- Centres de Recherche en Nutrition Humaine-Ouest, Nantes, France
| | - Juliana Guimaraes-Laguna
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Marie-Bernadette Maillard
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Eric Guédon
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Michel Neunlist
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| | - Yves Le Loir
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Gwénaël Jan
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Malvyne Rolli-Derkinderen
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| |
Collapse
|
4
|
Yung C, Zhang Y, Kuhn M, Armstrong RJ, Olyaei A, Aloia M, Scottoline B, Andres SF. Neonatal enteroids absorb extracellular vesicles from human milk-fed infant digestive fluid. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.03.556067. [PMID: 38187651 PMCID: PMC10769189 DOI: 10.1101/2023.09.03.556067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Human milk contains extracellular vesicles (HMEVs). Pre-clinical models suggest that HMEVs may enhance intestinal function and limit inflammation; however, it is unknown if HMEVs or their cargo survive neonatal human digestion. This limits the ability to leverage HMEV cargo as additives to infant nutrition or as therapeutics. This study aimed to develop an EV isolation pipeline from small volumes of human milk and neonatal intestinal contents after milk feeding (digesta) to address the hypothesis that HMEVs survive in vivo neonatal digestion to be taken up intestinal epithelial cells (IECs). Digesta was collected from nasoduodenal sampling tubes or ostomies. EVs were isolated from raw and pasteurized human milk and digesta by density-gradient ultracentrifugation following two-step skimming, acid precipitation of caseins, and multi-step filtration. EVs were validated by electron microscopy, western blotting, nanoparticle tracking analysis, resistive pulse sensing, and super-resolution microscopy. EV uptake was tested in human neonatal enteroids. HMEVs and digesta EVs (dEVs) show typical EV morphology and are enriched in CD81 and CD9, but depleted of β-casein and lactalbumin. HMEV and some dEV fractions contain mammary gland-derived protein BTN1A1. Neonatal human enteroids rapidly take up dEVs in part via clathrin-mediated endocytosis. Our data suggest that EVs can be isolated from digestive fluid and that these dEVs can be absorbed by IECs.
Collapse
|
5
|
Gurunathan S, Thangaraj P, Das J, Kim JH. Antibacterial and antibiofilm effects of Pseudomonas aeruginosa derived outer membrane vesicles against Streptococcus mutans. Heliyon 2023; 9:e22606. [PMID: 38125454 PMCID: PMC10730581 DOI: 10.1016/j.heliyon.2023.e22606] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023] Open
Abstract
Antimicrobial resistance (AMR) is a serious and most urgent global threat to human health. AMR is one of today's biggest difficulties in the health system and has the potential to harm people at any stage of life, making it a severe public health issue. There must be fewer antimicrobial medicines available to treat diseases given the rise in antibiotic-resistant organisms. If no new drugs are created or discovered, it is predicted that there won't be any effective antibiotics accessible by 2050. In most cases, Streptococcus increased antibiotic resistance by forming biofilms, which account for around 80 % of all microbial infections in humans. This highlights the need to look for new strategies to manage diseases that are resistant to antibiotics. Therefore, development alternative, biocompatible and high efficacy new strategies are essential to overcome drug resistance. Recently, bacterial derived extracellular vesicles have been applied to tackle infection and reduce the emergence of drug resistance. Therefore, the objective of the current study was designed to assess the antibacterial and antibiofilm potential of outer membrane vesicles (OMVs) derived from Pseudomonas aeruginosa againstStreptococcus mutans. According to the findings of this investigation, the pure P. aeruginosa outer membrane vesicles (PAOMVs) display a size of 100 nm. S. mutans treated with PAOMVs showed significant antibacterial and antibiofilm activity. The mechanistic studies revealed that PAOMVs induce cell death through excessive generation of reactive oxygen species and imbalance of redox leads to lipid peroxidation, decreased level of antioxidant markers including glutathione, superoxide dismutase and catalase. Further this study confirmed that PAOMVs significantly impairs metabolic activity through inhibiting lactate dehydrogenase activity (LDH), adenosine triphosphate (ATP) production, leakage of proteins and sugars. Interestingly, combination of sub-lethal concentrations of PAOMVs and antibiotics enhances cell death and biofilm formation of S. mutans. Altogether, this work, may serve as an important basis for further evaluation of PAOMVs as novel therapeutic agents against bacterial infections.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Biotechnology, Rathinam College of Arts and Science, Rathinam Techzone Campus, Eachanari, Coimbatore, 641 021, Tamil Nadu, India
| | - Pratheep Thangaraj
- Department of Biotechnology, Rathinam College of Arts and Science, Rathinam Techzone Campus, Eachanari, Coimbatore, 641 021, Tamil Nadu, India
| | - Joydeep Das
- Department of Chemistry, Mizoram University, Aizawl, 796 004, Mizoram, India
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, South Korea
| |
Collapse
|
6
|
Liu Z, Sokratian A, Duda AM, Xu E, Stanhope C, Fu A, Strader S, Li H, Yuan Y, Bobay BG, Sipe J, Bai K, Lundgaard I, Liu N, Hernandez B, Rickman CB, Miller SE, West AB. Anionic Nanoplastic Contaminants Promote Parkinson's Disease-Associated α-Synuclein Aggregation. RESEARCH SQUARE 2023:rs.3.rs-3439102. [PMID: 37886561 PMCID: PMC10602106 DOI: 10.21203/rs.3.rs-3439102/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Recent studies have identified increasing levels of nanoplastic pollution in the environment. Here we find that anionic nanoplastic contaminants potently precipitate the formation and propagation of α-synuclein protein fibrils through a high-affinity interaction with the amphipathic and non-amyloid component (NAC) domains in α-synuclein. Nanoplastics can internalize in neurons through clathrin-dependent endocytosis, causing a mild lysosomal impairment that slows the degradation of aggregated α-synuclein. In mice, nanoplastics combine with α-synuclein fibrils to exacerbate the spread of α-synuclein pathology across interconnected vulnerable brain regions, including the strong induction of α-synuclein inclusions in dopaminergic neurons in the substantia nigra. These results highlight a potential link for further exploration between nanoplastic pollution and α-synuclein aggregation associated with Parkinson's disease and related dementias.
Collapse
Affiliation(s)
- Zhiyong Liu
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
| | - Arpine Sokratian
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
| | | | - Enquan Xu
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
| | - Christina Stanhope
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
| | - Amber Fu
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
| | - Samuel Strader
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
| | - Huizhong Li
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
| | - Yuan Yuan
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
| | | | - Joana Sipe
- Department of Civil and Environmental Engineering, Duke University, Durham, NC, USA
| | - Ketty Bai
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
| | - Iben Lundgaard
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Na Liu
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Belinda Hernandez
- Department of Ophthalmology and Cell Biology, Duke University, Durham, NC, USA
| | | | - Sara E Miller
- Department of Pathology, Duke University, Durham, North Carolina, USA
| | - Andrew B. West
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| |
Collapse
|
7
|
Saadeldin IM, Bang S, Maigoro AY, Yun SH, Kim SI, Lee S, Cho J. Proteomic Analysis and Reprogramming Potential of the Porcine Intra-Ooplasmic Nanovesicles. Cell Reprogram 2023; 25:238-250. [PMID: 37725012 DOI: 10.1089/cell.2023.0050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023] Open
Abstract
Oocytes contain reprogramming machinery that can transform somatic cells into totipotent cells. In this study, we aimed to isolate and characterize nanovesicles from mature porcine oocytes and described them for the first time as "intra-ooplasmic vesicles (IOVs)". Isolated IOVs had an average diameter of 186.3 ± 10.8 nm. Proteomic analysis revealed 467 peptide reads, with the top 20 proteins related to reprogramming, antioxidative defense, cytoskeleton, heat shock proteins, and metabolism. Protein-protein interaction and gene ontology analysis indicated that these proteins were involved in various biological pathways, including protein folding, metabolism, and cellular responses to stress. Supplementing cultured fibroblasts with IOVs resulted in the expression of the pluripotency marker OCT4 and the early trophoblastic marker CDX2 and increased expression of the corresponding mRNAs together with increasing KLF4 and SALL4 expression. IOV treatment of fibroblasts for 14 consecutive days resulted in changes in cell morphology, with increased expression of ZEB2 and YBX3 as markers for epithelial-to-mesenchymal transition (EMT). These results provide a rationale for further characterization of IOVs, investigation of potential reprogramming capabilities for EMT, and the generation of induced pluripotent or oligopotent stem cells.
Collapse
Affiliation(s)
- Islam M Saadeldin
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- Research Institute of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Seonggyu Bang
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Abdulkadir Y Maigoro
- Department of Life Sciences, Incheon National University, Incheon, Republic of Korea
| | - Sung Ho Yun
- Korea Basic Science Institute (KBSI), Ochang, Republic of Korea
| | - Seung Ii Kim
- Korea Basic Science Institute (KBSI), Ochang, Republic of Korea
| | - Sanghoon Lee
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jongki Cho
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
8
|
Schöler D, Loosen SH, Wirtz TH, Brozat JF, dos Santos Ferreira Grani LA, Luedde T, Heinrichs L, Frank D, Koch A, Roderburg C, Spehlmann ME. Low extracellular vesicle concentrations predict survival in patients with heart failure. Front Cardiovasc Med 2023; 10:1163525. [PMID: 37293281 PMCID: PMC10244507 DOI: 10.3389/fcvm.2023.1163525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/11/2023] [Indexed: 06/10/2023] Open
Abstract
Background Heart disease is of worldwide importance due to high morbidity and mortality. Extracellular vesicle (EV) concentration and size represent novel diagnostic and prognostic biomarkers, e.g. in patients with liver cancer, but data on their prognostic relevance in heart disease are lacking. Here, we investigated the role of EV concentration, size and zeta potential in patients with heart disease. Methods Vesicle size distribution, concentration and zeta potential were measured by nanoparticle tracking analysis (NTA) in 28 intensive care unit (ICU) and 20 standard care (SC) patients and 20 healthy controls. Results Patients with any disease had a lower zeta potential compared to the healthy controls. Vesicle size (X50) was significantly higher in ICU patients (245 nm) with heart disease as compared to those patients with heart disease receiving standard care (195 nm), or healthy controls (215 nm) (p = 0.001). Notably, EV concentration was lower in ICU patients with heart disease (4.68 × 1010 particles/ml) compared to SC patients with heart disease (7,62 × 1010 particles/ml) and healthy controls (1.50 × 1011 particles/ml) (p = 0.002). Extracellular vesicle concentration is prognostic for overall survival in patients with heart disease. Overall survival is significantly reduced when the vesicle concentration is below 5.55 × 1010 particles/ml. Median overall survival was only 140 days in patients with vesicle concentrations below 5.55 × 1010 particles/ml compared to 211 days in patients with vesicle concentrations above 5.55 × 1010 particles/ml (p = 0.032). Summary Concentration of EVs is a novel prognostic marker in ICU and SC patients with heart disease.
Collapse
Affiliation(s)
- David Schöler
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sven H. Loosen
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Theresa H. Wirtz
- Department of Gastroenterology, Digestive Diseases and Intensive Care Medicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Jonathan F. Brozat
- Department of Gastroenterology, Digestive Diseases and Intensive Care Medicine, RWTH Aachen University Hospital, Aachen, Germany
- Department of Hepatology and Gastroenterology, Charite—Universitätsmedizin Berlin Campus Virchow-Klinikum (CVK) and Campus Charite Mitte (CCM), Berlin, Germany
| | - Lauredana A. dos Santos Ferreira Grani
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tom Luedde
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Lisa Heinrichs
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Derk Frank
- Internal Medicine III, University Hospital of Schleswig Holstein, Campus Kiel, Kiel, Germany
| | - Alexander Koch
- Department of Gastroenterology, Digestive Diseases and Intensive Care Medicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Christoph Roderburg
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Martina E. Spehlmann
- Internal Medicine III, University Hospital of Schleswig Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
9
|
Oviduct Epithelial Cell-Derived Extracellular Vesicles Improve Porcine Trophoblast Outgrowth. Vet Sci 2022; 9:vetsci9110609. [DOI: 10.3390/vetsci9110609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/20/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Porcine species have a great impact on studies on biomaterial production, organ transplantation and the development of biomedical models. The low efficiency of in vitro-produced embryos to derive embryonic stem cells has made achieving this goal a challenge. The fallopian tube plays an important role in the development of embryos. Extracellular vesicles (EVs) secreted by oviductal epithelial cells play an important role in the epigenetic regulation of embryo development. We used artificially isolated oviductal epithelial cells and EVs. In this study, oviductal epithelial cell (OEC) EVs were isolated and characterized through transmission electron microscopy, nanoparticles tracking analysis, western blotting and proteomics. We found that embryo development and blastocyst formation rate was significantly increased (14.3% ± 0.6% vs. 6.0% ± 0.6%) after OEC EVs treatment. According to our data, the inner cell mass (ICM)/trophectoderm (TE) ratio of the embryonic cell number increased significantly after OEC EVs treatment (43.7% ± 2.3% vs. 28.4% ± 2.1%). Meanwhile, the attachment ability of embryos treated with OEV EVs was significantly improved (43.5% ± 2.1% vs. 29.2% ± 2.5%, respectively). Using quantitative polymerase chain reaction (qPCR), we found that the expression of reprogramming genes (POU5F1, SOX2, NANOG, KLF4 and c-Myc) and implantation-related genes (VIM, KRT8, TEAD4 and CDX2) significantly increased in OEC EV-treated embryos. We report that OEC EV treatment can improve the development and implantation abilities of embryos.
Collapse
|
10
|
Saadeldin IM, Tanga BM, Bang S, Seo C, Koo O, Yun SH, Kim SI, Lee S, Cho J. ROCK Inhibitor (Y-27632) Abolishes the Negative Impacts of miR-155 in the Endometrium-Derived Extracellular Vesicles and Supports Embryo Attachment. Cells 2022; 11:cells11193178. [PMID: 36231141 PMCID: PMC9564368 DOI: 10.3390/cells11193178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Extracellular vesicles (EVs) are nanosized vesicles that act as snapshots of cellular components and mediate cellular communications, but they may contain cargo contents with undesired effects. We developed a model to improve the effects of endometrium-derived EVs (Endo-EVs) on the porcine embryo attachment in feeder-free culture conditions. Endo-EVs cargo contents were analyzed using conventional and real-time PCR for micro-RNAs, messenger RNAs, and proteomics. Porcine embryos were generated by parthenogenetic electric activation in feeder-free culture conditions supplemented with or without Endo-EVs. The cellular uptake of Endo-EVs was confirmed using the lipophilic dye PKH26. Endo-EVs cargo contained miR-100, miR-132, and miR-155, together with the mRNAs of porcine endogenous retrovirus (PERV) and β-catenin. Targeting PERV with CRISPR/Cas9 resulted in reduced expression of PERV mRNA transcripts and increased miR-155 in the Endo-EVs, and supplementing these in embryos reduced embryo attachment. Supplementing the medium containing Endo-EVs with miR-155 inhibitor significantly improved the embryo attachment with a few outgrowths, while supplementing with Rho-kinase inhibitor (RI, Y-27632) dramatically improved both embryo attachment and outgrowths. Moreover, the expression of miR-100, miR-132, and the mRNA transcripts of BCL2, zinc finger E-box-binding homeobox 1, β-catenin, interferon-γ, protein tyrosine phosphatase non-receptor type 1, PERV, and cyclin-dependent kinase 2 were all increased in embryos supplemented with Endo-EVs + RI compared to those in the control group. Endo-EVs + RI reduced apoptosis and increased the expression of OCT4 and CDX2 and the cell number of embryonic outgrowths. We examined the individual and combined effects of RI compared to those of the miR-155 mimic and found that RI can alleviate the negative effects of the miR-155 mimic on embryo attachment and outgrowths. EVs can improve embryo attachment and the unwanted effects of the de trop cargo contents (miR-155) can be alleviated through anti-apoptotic molecules such as the ROCK inhibitor.
Collapse
Affiliation(s)
- Islam M. Saadeldin
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
- Research Institute of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Bereket Molla Tanga
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Seonggyu Bang
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Chaerim Seo
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | | | - Sung Ho Yun
- Korea Basic Science Institute (KBSI), Ochang 28119, Korea
| | - Seung Il Kim
- Korea Basic Science Institute (KBSI), Ochang 28119, Korea
| | - Sanghoon Lee
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Jongki Cho
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
- Correspondence: ; Tel.: +82-42-821-6788
| |
Collapse
|
11
|
Manukonda R, Yenuganti VR, Nagar N, Dholaniya PS, Malpotra S, Attem J, Reddy MM, Jakati S, Mishra DK, Reddanna P, Poluri KM, Vemuganti GK, Kaliki S. Comprehensive Analysis of Serum Small Extracellular Vesicles-Derived Coding and Non-Coding RNAs from Retinoblastoma Patients for Identifying Regulatory Interactions. Cancers (Basel) 2022; 14:cancers14174179. [PMID: 36077715 PMCID: PMC9454787 DOI: 10.3390/cancers14174179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/18/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
The present study employed nanoparticle tracking analysis, transmission electron microscopy, immunoblotting, RNA sequencing, and quantitative real-time PCR validation to characterize serum-derived small extracellular vesicles (sEVs) from RB patients and age-matched controls. Bioinformatics methods were used to analyze functions, and regulatory interactions between coding and non-coding (nc) sEVs RNAs. The results revealed that the isolated sEVs are round-shaped with a size < 150 nm, 5.3 × 1011 ± 8.1 particles/mL, and zeta potential of 11.1 to −15.8 mV, and expressed exosome markers CD9, CD81, and TSG101. A total of 6514 differentially expressed (DE) mRNAs, 123 DE miRNAs, and 3634 DE lncRNAs were detected. Both miRNA-mRNA and lncRNA-miRNA-mRNA network analysis revealed that the cell cycle-specific genes including CDKNI1A, CCND1, c-MYC, and HIF1A are regulated by hub ncRNAs MALAT1, AFAP1-AS1, miR145, 101, and 16-5p. Protein-protein interaction network analysis showed that eye-related DE mRNAs are involved in rod cell differentiation, cone cell development, and retinol metabolism. In conclusion, our study provides a comprehensive overview of the RB sEV RNAs and regulatory interactions between them.
Collapse
Affiliation(s)
- Radhika Manukonda
- The Operation Eyesight Universal Institute for Eye Cancer, L V Prasad Eye Institute, Hyderabad 500034, India
- Brien Holden Eye Research Center, L V Prasad Eye Institute, Hyderabad 500034, India
| | - Vengala Rao Yenuganti
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Prof. C.R. Rao Road, Gachibowli, Hyderabad 500046, India or
| | - Nupur Nagar
- Department of Biosciences and Bioengineering, Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Pankaj Singh Dholaniya
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Prof. C.R. Rao Road, Gachibowli, Hyderabad 500046, India
| | - Shivani Malpotra
- The Operation Eyesight Universal Institute for Eye Cancer, L V Prasad Eye Institute, Hyderabad 500034, India
- Brien Holden Eye Research Center, L V Prasad Eye Institute, Hyderabad 500034, India
| | - Jyothi Attem
- School of Medical Sciences, Science Complex, University of Hyderabad, Prof. C.R. Rao Road, Gachibowli, Hyderabad 500046, India
| | - Mamatha M. Reddy
- The Operation Eyesight Universal Institute for Eye Cancer, L V Prasad Eye Institute, Bhubaneswar 751024, India or
| | - Saumya Jakati
- Ophthalmic Pathology Laboratory, L V Prasad Eye Institute, Hyderabad 500034, India
| | - Dilip K Mishra
- Ophthalmic Pathology Laboratory, L V Prasad Eye Institute, Hyderabad 500034, India
| | - Pallu Reddanna
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Prof. C.R. Rao Road, Gachibowli, Hyderabad 500046, India or
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering, Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Geeta K. Vemuganti
- School of Medical Sciences, Science Complex, University of Hyderabad, Prof. C.R. Rao Road, Gachibowli, Hyderabad 500046, India
| | - Swathi Kaliki
- The Operation Eyesight Universal Institute for Eye Cancer, L V Prasad Eye Institute, Hyderabad 500034, India
- Correspondence: ; Tel.: +91-40-68102502
| |
Collapse
|
12
|
Kis D, Csordás IB, Persa E, Jezsó B, Hargitai R, Szatmári T, Sándor N, Kis E, Balázs K, Sáfrány G, Lumniczky K. Extracellular Vesicles Derived from Bone Marrow in an Early Stage of Ionizing Radiation Damage Are Able to Induce Bystander Responses in the Bone Marrow. Cells 2022; 11:cells11010155. [PMID: 35011718 PMCID: PMC8750882 DOI: 10.3390/cells11010155] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 02/01/2023] Open
Abstract
Ionizing radiation (IR)-induced bystander effects contribute to biological responses to radiation, and extracellular vesicles (EVs) play important roles in mediating these effects. In this study we investigated the role of bone marrow (BM)-derived EVs in the bystander transfer of radiation damage. Mice were irradiated with 0.1Gy, 0.25Gy and 2Gy, EVs were extracted from the BM supernatant 24 h or 3 months after irradiation and injected into bystander mice. Acute effects on directly irradiated or EV-treated mice were investigated after 4 and 24 h, while late effects were investigated 3 months after treatment. The acute effects of EVs on the hematopoietic stem and progenitor cell pools were similar to direct irradiation effects and persisted for up to 3 months, with the hematopoietic stem cells showing the strongest bystander responses. EVs isolated 3 months after irradiation elicited no bystander responses. The level of seven microRNAs (miR-33a-3p, miR-140-3p, miR-152-3p, miR-199a-5p, miR-200c-5p, miR-375-3p and miR-669o-5p) was altered in the EVs isolated 24 hour but not 3 months after irradiation. They regulated pathways highly relevant for the cellular response to IR, indicating their role in EV-mediated bystander responses. In conclusion, we showed that only EVs from an early stage of radiation damage could transmit IR-induced bystander effects.
Collapse
Affiliation(s)
- Dávid Kis
- National Public Health Center, Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, 1097 Budapest, Hungary; (D.K.); (I.B.C.); (E.P.); (R.H.); (T.S.); (N.S.); (E.K.); (K.B.); (G.S.)
- Doctoral School of Pathological Sciences, Semmelweis University, 1085 Budapest, Hungary
| | - Ilona Barbara Csordás
- National Public Health Center, Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, 1097 Budapest, Hungary; (D.K.); (I.B.C.); (E.P.); (R.H.); (T.S.); (N.S.); (E.K.); (K.B.); (G.S.)
| | - Eszter Persa
- National Public Health Center, Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, 1097 Budapest, Hungary; (D.K.); (I.B.C.); (E.P.); (R.H.); (T.S.); (N.S.); (E.K.); (K.B.); (G.S.)
| | - Bálint Jezsó
- Doctoral School of Biology and Institute of Biology, Eötvös Loránd University, 1053 Budapest, Hungary;
- Research Centre for Natural Sciences, Institute of Enzymology, 1117 Budapest, Hungary
| | - Rita Hargitai
- National Public Health Center, Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, 1097 Budapest, Hungary; (D.K.); (I.B.C.); (E.P.); (R.H.); (T.S.); (N.S.); (E.K.); (K.B.); (G.S.)
| | - Tünde Szatmári
- National Public Health Center, Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, 1097 Budapest, Hungary; (D.K.); (I.B.C.); (E.P.); (R.H.); (T.S.); (N.S.); (E.K.); (K.B.); (G.S.)
| | - Nikolett Sándor
- National Public Health Center, Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, 1097 Budapest, Hungary; (D.K.); (I.B.C.); (E.P.); (R.H.); (T.S.); (N.S.); (E.K.); (K.B.); (G.S.)
| | - Enikő Kis
- National Public Health Center, Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, 1097 Budapest, Hungary; (D.K.); (I.B.C.); (E.P.); (R.H.); (T.S.); (N.S.); (E.K.); (K.B.); (G.S.)
| | - Katalin Balázs
- National Public Health Center, Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, 1097 Budapest, Hungary; (D.K.); (I.B.C.); (E.P.); (R.H.); (T.S.); (N.S.); (E.K.); (K.B.); (G.S.)
- Doctoral School of Pathological Sciences, Semmelweis University, 1085 Budapest, Hungary
| | - Géza Sáfrány
- National Public Health Center, Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, 1097 Budapest, Hungary; (D.K.); (I.B.C.); (E.P.); (R.H.); (T.S.); (N.S.); (E.K.); (K.B.); (G.S.)
| | - Katalin Lumniczky
- National Public Health Center, Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, 1097 Budapest, Hungary; (D.K.); (I.B.C.); (E.P.); (R.H.); (T.S.); (N.S.); (E.K.); (K.B.); (G.S.)
- Correspondence:
| |
Collapse
|
13
|
Soltész B, Buglyó G, Németh N, Szilágyi M, Pös O, Szemes T, Balogh I, Nagy B. The Role of Exosomes in Cancer Progression. Int J Mol Sci 2021; 23:ijms23010008. [PMID: 35008434 PMCID: PMC8744561 DOI: 10.3390/ijms23010008] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/05/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022] Open
Abstract
Early detection, characterization and monitoring of cancer are possible by using extracellular vesicles (EVs) isolated from non-invasively obtained liquid biopsy samples. They play a role in intercellular communication contributing to cell growth, differentiation and survival, thereby affecting the formation of tumor microenvironments and causing metastases. EVs were discovered more than seventy years ago. They have been tested recently as tools of drug delivery to treat cancer. Here we give a brief review on extracellular vesicles, exosomes, microvesicles and apoptotic bodies. Exosomes play an important role by carrying extracellular nucleic acids (DNA, RNA) in cell-to-cell communication causing tumor and metastasis development. We discuss the role of extracellular vesicles in the pathogenesis of cancer and their practical application in the early diagnosis, follow up, and next-generation treatment of cancer patients.
Collapse
Affiliation(s)
- Beáta Soltész
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (G.B.); (N.N.); (M.S.); (I.B.); (B.N.)
- Correspondence: ; Tel.: +36-52416531
| | - Gergely Buglyó
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (G.B.); (N.N.); (M.S.); (I.B.); (B.N.)
| | - Nikolett Németh
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (G.B.); (N.N.); (M.S.); (I.B.); (B.N.)
| | - Melinda Szilágyi
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (G.B.); (N.N.); (M.S.); (I.B.); (B.N.)
| | - Ondrej Pös
- Geneton Ltd., 841 04 Bratislava, Slovakia; (O.P.); (T.S.)
- Comenius University Science Park, Comenius University, 841 04 Bratislava, Slovakia
| | - Tomas Szemes
- Geneton Ltd., 841 04 Bratislava, Slovakia; (O.P.); (T.S.)
- Comenius University Science Park, Comenius University, 841 04 Bratislava, Slovakia
| | - István Balogh
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (G.B.); (N.N.); (M.S.); (I.B.); (B.N.)
- Division of Clinical Genetics, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Bálint Nagy
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (G.B.); (N.N.); (M.S.); (I.B.); (B.N.)
| |
Collapse
|
14
|
Azevedo CAB, da Cunha RS, Junho CVC, da Silva JV, Moreno-Amaral AN, de Moraes TP, Carneiro-Ramos MS, Stinghen AEM. Extracellular Vesicles and Their Relationship with the Heart-Kidney Axis, Uremia and Peritoneal Dialysis. Toxins (Basel) 2021; 13:toxins13110778. [PMID: 34822562 PMCID: PMC8618757 DOI: 10.3390/toxins13110778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/19/2021] [Accepted: 10/28/2021] [Indexed: 12/18/2022] Open
Abstract
Cardiorenal syndrome (CRS) is described as primary dysfunction in the heart culminating in renal injury or vice versa. CRS can be classified into five groups, and uremic toxin (UT) accumulation is observed in all types of CRS. Protein-bound uremic toxin (PBUT) accumulation is responsible for permanent damage to the renal tissue, and mainly occurs in CRS types 3 and 4, thus compromising renal function directly leading to a reduction in the glomerular filtration rate (GFR) and/or subsequent proteinuria. With this decrease in GFR, patients may need renal replacement therapy (RRT), such as peritoneal dialysis (PD). PD is a high-quality and home-based dialysis therapy for patients with end-stage renal disease (ESRD) and is based on the semi-permeable characteristics of the peritoneum. These patients are exposed to factors which may cause several modifications on the peritoneal membrane. The presence of UT may harm the peritoneum membrane, which in turn can lead to the formation of extracellular vesicles (EVs). EVs are released by almost all cell types and contain lipids, nucleic acids, metabolites, membrane proteins, and cytosolic components from their cell origin. Our research group previously demonstrated that the EVs can be related to endothelial dysfunction and are formed when UTs are in contact with the endothelial monolayer. In this scenario, this review explores the mechanisms of EV formation in CRS, uremia, the peritoneum, and as potential biomarkers in peritoneal dialysis.
Collapse
Affiliation(s)
- Carolina Amaral Bueno Azevedo
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba 81531-980, Brazil; (C.A.B.A.); (R.S.d.C.)
| | - Regiane Stafim da Cunha
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba 81531-980, Brazil; (C.A.B.A.); (R.S.d.C.)
| | - Carolina Victoria Cruz Junho
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André 09210-580, Brazil; (C.V.C.J.); (J.V.d.S.); (M.S.C.-R.)
| | - Jessica Verônica da Silva
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André 09210-580, Brazil; (C.V.C.J.); (J.V.d.S.); (M.S.C.-R.)
| | - Andréa N. Moreno-Amaral
- Graduate Program in Health Sciences, School of Medicine, Pontifical Catholic University of Paraná, Curitiba 80215-901, Brazil; (A.N.M.-A.); (T.P.d.M.)
| | - Thyago Proença de Moraes
- Graduate Program in Health Sciences, School of Medicine, Pontifical Catholic University of Paraná, Curitiba 80215-901, Brazil; (A.N.M.-A.); (T.P.d.M.)
| | - Marcela Sorelli Carneiro-Ramos
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André 09210-580, Brazil; (C.V.C.J.); (J.V.d.S.); (M.S.C.-R.)
| | - Andréa Emilia Marques Stinghen
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba 81531-980, Brazil; (C.A.B.A.); (R.S.d.C.)
- Correspondence:
| |
Collapse
|
15
|
Schöler D, Castoldi M, Jördens MS, Schulze-Hagen M, Kuhl C, Keitel V, Luedde T, Roderburg C, Loosen SH. Enlarged extracellular vesicles are a negative prognostic factor in patients undergoing TACE for primary or secondary liver cancer-a case series. PLoS One 2021; 16:e0255983. [PMID: 34407090 PMCID: PMC8372935 DOI: 10.1371/journal.pone.0255983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 07/27/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Transarterial chemoembolization (TACE) has evolved as a standard treatment option in patients with intermediate stage, unresectable HCC [Barcelona Clinic Liver Cancer (BCLC) stage B] as well as in patients with liver metastases, when surgery or systemic therapy is considered not appropriate. Concentration and sizes of extracellular vesicles (EVs) recently emerged as novel diagnostic and prognostic biomarkers in patients with liver cancer, but no data on its prognostic relevance in the context of TACE exists. Here, we evaluate pre-interventional EVs as a potential biomarker in patients undergoing TACE for primary and secondary hepatic malignancies. METHODS Vesicle size distribution and concentration were measured by nanoparticle tracking analysis (NTA) in patient sera before and after TACE in 38 patients. RESULTS Extracellular vesicle size distribution measured before TACE is of prognostic significance with respect to overall survival in patients after TACE. Overall survival is significantly reduced when initial vesicle size (X50) is in the upper quartile (>145.65nm). Median overall survival in patients in the upper quartile was only 314 days, compared to 799 days in patients with vesicle size in the first to third quartile (<145.65nm; p = 0.007). Vesicle size was also shown to be a significant prognostic marker for overall survival in Cox regression analysis [HR 1.089, 95% CI: 1.021-1.162, p = 0.010]. In addition, a significant correlation was observed between initial EVs concentration/BMI (rS = 0.358, p = 0.029), X50/IL-8-concentration (rS = 0.409, p = 0.011) and X50/CRP-concentration (rS = 0.404, p = 0.016). In contrast, with regard to immediate tumor response after TACE, EVs concentration and size did not differ. SUMMARY Sizes (but not concentrations) of EVs represent a novel prognostic marker in patients receiving TACE for primary and secondary hepatic malignancies since patients with enlarged EVs display a significantly impaired prognosis after TACE.
Collapse
Affiliation(s)
- David Schöler
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Mirco Castoldi
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Markus S. Jördens
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Max Schulze-Hagen
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Christiane Kuhl
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Verena Keitel
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tom Luedde
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christoph Roderburg
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sven H. Loosen
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
16
|
Abstract
Extracellular Vesicles (EVs) gained significant interest within the last decade as a new source of biomarkers for the early detection of diseases and a promising tool for therapeutic applications. In this work, we present Extracellular Vesicles Quantitative Capillary Electrophoresis (EVqCE) to measure an average mass of RNA in EVs, determine EV concentrations and the degree of EV degradation after sample handling. We used EVqCE to analyze EVs isolated from conditioned media of three cancer cell lines. EVqCE employs capillary zone electrophoresis with laser-induced fluorescent detection to separate intact EVs from free nucleic acids. After lysis of EVs with a detergent, the encapsulated nucleic acids are released. Therefore, the initial concentration of intact EVs is calculated based on a nucleic acid peak gain. EVqCE works in a dynamic range of EV concentrations from 108 to 1010 particles/mL. The quantification process can be completed in less than one hour and requires minimum optimization. Furthermore, the average mass of RNA was found to be in the range of 200–400 ag per particle, noting that more aggressive cancer cells have less RNA in EVs (200 ag per particle) than non-aggressive cancer cells (350 ag per particle). EVqCE works well for the degradation analysis of EVs. Sonication for 10 min at 40 kHz caused 85% degradation of EVs, 10 freeze-thaw cycles (from −80 °C to 22 °C) produced 40%, 14-day storage at 4 °C made 32%, and vortexing for 5 min caused 5% degradation. Presently, EVqCE cannot separate and distinguish individual EV populations (exosomes, microvesicles, apoptotic bodies) from each other. Still, it is tolerant to the presence of non-EV particles, protein-lipid complexes, and protein aggregates.
Collapse
|
17
|
Effects of Extracellular Vesicles from Blood-Derived Products on Osteoarthritic Chondrocytes within an Inflammation Model. Int J Mol Sci 2021; 22:ijms22137224. [PMID: 34281278 PMCID: PMC8267849 DOI: 10.3390/ijms22137224] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is hallmarked by a progressive degradation of articular cartilage. One major driver of OA is inflammation, in which cytokines such as IL-6, TNF-α and IL-1β are secreted by activated chondrocytes, as well as synovial cells—including macrophages. Intra-articular injection of blood products—such as citrate-anticoagulated plasma (CPRP), hyperacute serum (hypACT), and extracellular vesicles (EVs) isolated from blood products—is gaining increasing importance in regenerative medicine for the treatment of OA. A co-culture system of primary OA chondrocytes and activated M1 macrophages was developed to model an OA joint in order to observe the effects of EVs in modulating the inflammatory environment. Primary OA chondrocytes were obtained from patients undergoing total knee replacement. Primary monocytes obtained from voluntary healthy donors and the monocytic cell line THP-1 were differentiated and activated into proinflammatory M1 macrophages. EVs were isolated by ultracentrifugation and characterized by nanoparticle tracking analysis and Western blot. Gene expression analysis of chondrocytes by RT-qPCR revealed increased type II collagen expression, while cytokine profiling via ELISA showed lower TNF-α and IL-1β levels associated with EV treatment. In conclusion, the inflammation model provides an accessible tool to investigate the effects of blood products and EVs in the inflammatory context of OA.
Collapse
|
18
|
Shen Y, Wang L, Wu Y, Ou Y, Lu H, Yao X. A novel diagnostic signature based on three circulating exosomal mircoRNAs for chronic obstructive pulmonary disease. Exp Ther Med 2021; 22:717. [PMID: 34007326 PMCID: PMC8120666 DOI: 10.3892/etm.2021.10149] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 03/18/2021] [Indexed: 12/15/2022] Open
Abstract
Exosomal microRNAs (exo-miRNAs or miRs) have demonstrated diagnostic value in various diseases. However, their diagnostic value in chronic obstructive pulmonary disease (COPD) has yet to be fully established. The purpose of the present study was to screen differentially expressed exo-miRNAs in the plasma of patients with COPD and healthy individuals and to evaluate their potential diagnostic value in COPD. Differentially expressed exo-miRNAs in the plasma of patients with COPD and controls were identified using high-throughput sequencing and confirmed using reverse transcription-quantitative PCR (RT-qPCR). Bioinformatics analysis was then performed to predict the function of the selected exo-miRNAs and their target genes in COPD. After a network model was constructed, linear regression analysis was performed to determine the association between exo-miRNA expression and the clinical characteristics of subjects in a validated cohort (46 COPD cases; 34 matched healthy controls). Receiver operating characteristic curve was subsequently plotted to test the diagnostic value of the candidate biomarkers. The top 20 significantly aberrantly expressed COPD-associated exo-miRNAs were verified using RT-qPCR. Of these, nine were then selected for subsequent analysis, five of which were found to be upregulated (miR-23a, miR-1, miR-574, miR-152 and miR-221) and four of which were downregulated (miR-3158, miR-7706, miR-685 and miR-144). The results of Gene Ontology and KEGG pathway analysis revealed that these miRNAs were mainly involved in certain biological functions, such as metabolic processes, such as galactose metabolism and signaling pathways (PI3K-AKT) associated with COPD. The expression levels of three exo-miRNAs (miR-23a, miR-221 and miR-574) were found to be negatively associated with the forced expiratory volume in the 1st second/forced vital capacity. Furthermore, the area under the curve values of the three exo-miRNAs (miR-23a, miR-221 and miR-574) for COPD diagnosis were 0.776 [95% confidence interval (CI), 0.669-0.882], 0.688 (95% CI, 0.563-0.812) and 0.842 (95% CI, 0.752-0.931), respectively. In conclusion, the three circulating exosomal miRNAs (miR-23a, miR-221 and miR-574) may serve as novel circulating biomarkers for the diagnosis of COPD. These results may also enhance our understanding and provide novel potential treatment options for patients with COPD.
Collapse
Affiliation(s)
- Yahui Shen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
- Department of Respiratory and Critical Care Medicine, Taizhou Clinical Medical School of Nanjing Medical University, Taizhou, Jiangsu 225300, P.R. China
| | - Lina Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yunhui Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yingwei Ou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Huiyu Lu
- Department of Respiratory and Critical Care Medicine, Taizhou Clinical Medical School of Nanjing Medical University, Taizhou, Jiangsu 225300, P.R. China
| | - Xin Yao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
19
|
Mohammad S, Hutchinson KA, da Silva DF, Bhattacharjee J, McInnis K, Burger D, Adamo KB. Circulating small extracellular vesicles increase after an acute bout of moderate-intensity exercise in pregnant compared to non-pregnant women. Sci Rep 2021; 11:12615. [PMID: 34135428 PMCID: PMC8209031 DOI: 10.1038/s41598-021-92180-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
The physiological and molecular mechanisms linking prenatal physical activity and improvements in maternal–fetal health are unknown. It is hypothesized that small extracellular vesicles (EVs, ~ 10–120 nm) are involved in tissue cross-talk during exercise. We aimed to characterize the circulating small EV profile of pregnant versus non-pregnant women after an acute bout of moderate-intensity exercise. Pregnant (N = 10) and non-pregnant control (N = 9) women performed a single session of moderate-intensity treadmill walking for 30 min. Plasma was collected immediately pre- and post-exercise, and small EVs were isolated by differential ultracentrifugation. EV presence was confirmed by western blotting for the small EV proteins TSG-101 and flottilin-1. Small EVs were quantified by size and concentration using nanoparticle tracking analysis and transmission electron microscopy. All EV fractions were positive for TSG-101 and flotillin-1, and negative for calnexin. Mean vesicle size at baseline and percent change in size post-exercise were not different between groups. At baseline, pregnant women had higher levels of small EVs compared to controls (1.83E+10 ± 1.25E+10 particles/mL vs. 8.11E+09 ± 4.04E+09 particles/mL, respectively; p = 0.032). Post-exercise, small EVs increased significantly in the circulation of pregnant compared to non-pregnant women after correcting for baseline values (64.7 ± 24.6% vs. − 23.3 ± 26.1%, respectively; F = 5.305, p = 0.035). Further research is needed to assess the functional roles of exercise-induced small EVs in pregnancy.
Collapse
Affiliation(s)
- Shuhiba Mohammad
- Faculty of Health Sciences, School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada
| | - Kelly Ann Hutchinson
- Faculty of Health Sciences, School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada
| | | | - Jayonta Bhattacharjee
- Faculty of Health Sciences, School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada
| | - Kurt McInnis
- Faculty of Health Sciences, School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada
| | - Dylan Burger
- Kidney Research Centre, Department of Cellular and Molecular Medicine, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Kristi B Adamo
- Faculty of Health Sciences, School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada. .,, 200 Lees Avenue, Building E, Room E250F, Ottawa, ON, K1N 6N5, Canada.
| |
Collapse
|
20
|
Sjöberg M, Mapar M, Armanious A, Zhdanov VP, Agnarsson B, Höök F. Time-Resolved and Label-Free Evanescent Light-Scattering Microscopy for Mass Quantification of Protein Binding to Single Lipid Vesicles. NANO LETTERS 2021; 21:4622-4628. [PMID: 34003003 PMCID: PMC8289281 DOI: 10.1021/acs.nanolett.1c00644] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/05/2021] [Indexed: 06/12/2023]
Abstract
In-depth understanding of the intricate interactions between biomolecules and nanoparticles is hampered by a lack of analytical methods providing quantitative information about binding kinetics. Herein, we demonstrate how label-free evanescent light-scattering microscopy can be used to temporally resolve specific protein binding to individual surface-bound (∼100 nm) lipid vesicles. A theoretical model is proposed that translates protein-induced changes in light-scattering intensity into bound mass. Since the analysis is centered on individual lipid vesicles, the signal from nonspecific protein binding to the surrounding surface is completely avoided, offering a key advantage over conventional surface-based techniques. Further, by averaging the intensities from less than 2000 lipid vesicles, the sensitivity is shown to increase by orders of magnitude. Taken together, these features provide a new avenue in studies of protein-nanoparticle interaction, in general, and specifically in the context of nanoparticles in medical diagnostics and drug delivery.
Collapse
Affiliation(s)
- Mattias Sjöberg
- Division
of Nano and Biophysics, Department of Physics, Chalmers University of Technology, Gothenburg 41296, Sweden
| | - Mokhtar Mapar
- Division
of Nano and Biophysics, Department of Physics, Chalmers University of Technology, Gothenburg 41296, Sweden
| | - Antonius Armanious
- Division
of Nano and Biophysics, Department of Physics, Chalmers University of Technology, Gothenburg 41296, Sweden
| | - Vladimir P. Zhdanov
- Division
of Nano and Biophysics, Department of Physics, Chalmers University of Technology, Gothenburg 41296, Sweden
- Boreskov
Institute of Catalysis, Russian Academy
of Sciences, Novosibirsk 630090, Russia
| | - Björn Agnarsson
- Division
of Nano and Biophysics, Department of Physics, Chalmers University of Technology, Gothenburg 41296, Sweden
| | - Fredrik Höök
- Division
of Nano and Biophysics, Department of Physics, Chalmers University of Technology, Gothenburg 41296, Sweden
| |
Collapse
|
21
|
Han Z, Liu J, Liu Z, Pan W, Yang Y, Chen X, Gao Y, Duan X. Resistive pulse sensing device with embedded nanochannel (nanochannel-RPS) for label-free biomolecule and bionanoparticle analysis. NANOTECHNOLOGY 2021; 32:295507. [PMID: 33823494 DOI: 10.1088/1361-6528/abf510] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/06/2021] [Indexed: 06/12/2023]
Abstract
This paper reports an IC-compatible method for fabricating a PDMS-based resistive pulse sensing (RPS) device with embedded nanochannel (nanochannel-RPS) for label-free analysis of biomolecules and bionanoparticles, such as plasmid DNAs and exosomes. Here, a multilayer lithography process was proposed to fabricate the PDMS mold for the microfluidic device, comprising a bridging nanochannel, as the sensing gate. RPS was performed by placing the sensing and excitation electrodes symmetrically upstream and downstream of the sensing gate. In order to reduce the noise level, a reference electrode was designed and placed beside the excitation electrode. To demonstrate the feasibility of the proposed nanochannel-RPS device and sensing system, polystyrene micro- and nanoparticles with diameters of 1μm and 300 nm were tested by the proposed device with signal-to-noise ratios (SNR) ranging from 9.1-30.5 and 2.2-5.9, respectively. Furthermore, a nanochannel with height of 300 nm was applied for 4 kb plasmid DNA detection, implying the potential of the proposed method for label-free quantification of nanoscale biomolecules. Moreover, HeLa cell exosomes, known as a well-studied subtype of extracellular vesicles, were measured and analyzed by their size distribution. The result of the resistive pulse amplitude corresponded well to that of nanoparticle tracking analysis (NTA). The proposed nanochannel-RPS device and the sensing strategy are not only capable of label-free analysis for nanoscale biomolecules and bionanoparticles, but are also cost-effective for large-scale manufacturing.
Collapse
Affiliation(s)
- Ziyu Han
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instrument and Opto-electronics Engineering, Tianjin University, Tianjin 300072, People's Republic of China
| | - Jiantao Liu
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instrument and Opto-electronics Engineering, Tianjin University, Tianjin 300072, People's Republic of China
- Center for Advanced Measurement Science, National Institute of Metrology, Beijing 100029, People's Republic of China
| | - Zhanning Liu
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instrument and Opto-electronics Engineering, Tianjin University, Tianjin 300072, People's Republic of China
- Center for Advanced Measurement Science, National Institute of Metrology, Beijing 100029, People's Republic of China
| | - Wenwei Pan
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instrument and Opto-electronics Engineering, Tianjin University, Tianjin 300072, People's Republic of China
| | - Yang Yang
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instrument and Opto-electronics Engineering, Tianjin University, Tianjin 300072, People's Republic of China
| | - Xuejiao Chen
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instrument and Opto-electronics Engineering, Tianjin University, Tianjin 300072, People's Republic of China
| | - Yunhua Gao
- Center for Advanced Measurement Science, National Institute of Metrology, Beijing 100029, People's Republic of China
| | - Xuexin Duan
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instrument and Opto-electronics Engineering, Tianjin University, Tianjin 300072, People's Republic of China
| |
Collapse
|
22
|
Otahal A, Kuten-Pella O, Kramer K, Neubauer M, Lacza Z, Nehrer S, De Luna A. Functional repertoire of EV-associated miRNA profiles after lipoprotein depletion via ultracentrifugation and size exclusion chromatography from autologous blood products. Sci Rep 2021; 11:5823. [PMID: 33712660 PMCID: PMC7955123 DOI: 10.1038/s41598-021-84234-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/11/2021] [Indexed: 12/14/2022] Open
Abstract
Cartilage breakdown, inflammation and pain are hallmark symptoms of osteoarthritis, and autologous blood products such as citrate-anticoagulated platelet-rich plasma (CPRP) or hyperacute serum (hypACT) have been developed as a regenerative approach to rebuild cartilage, inhibit inflammation and reduce pain. However, mechanisms of action of these blood derivatives are still not fully understood, in part due to the large number of components present in these medical products. In addition, the discovery of extracellular vesicles (EVs) and their involvement in intercellular communication mediated by cargo molecules like microRNAs (miRNAs) opened up a whole new level of complexity in understanding blood products. In this study we focused on the development of an isolation protocol for EVs from CPRP and hypACT that can also deplete lipoproteins, which are often co-isolated in EV research due to shared physical properties. Several isolation methods were compared in terms of particle yield from CPRP and hypACT. To gain insights into the functional repertoire conveyed via EV-associated miRNAs, we performed functional enrichment analysis and identified NFκB signaling strongly targeted by CPRP EV miRNAs, whereas hypACT EV miRNAs affect IL6- and TGFβ/SMAD signaling.
Collapse
Affiliation(s)
- Alexander Otahal
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria
| | | | - Karina Kramer
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria
| | - Markus Neubauer
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria
| | - Zsombor Lacza
- Department of Sports Physiology, University of Physical Education, Budapest, Hungary
| | - Stefan Nehrer
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria
| | - Andrea De Luna
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria.
| |
Collapse
|
23
|
Mallach A, Gobom J, Zetterberg H, Hardy J, Piers TM, Wray S, Pocock JM. The influence of the R47H triggering receptor expressed on myeloid cells 2 variant on microglial exosome profiles. Brain Commun 2021; 3:fcab009. [PMID: 34704019 PMCID: PMC8244649 DOI: 10.1093/braincomms/fcab009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 01/08/2023] Open
Abstract
Variants in the triggering receptor expressed on myeloid cells 2 gene are linked with an increased risk of dementia, in particular the R47Hhet triggering receptor expressed on myeloid cells 2 variant is linked to late-onset Alzheimer's disease. Using human induced pluripotent stem cells-derived microglia, we assessed whether variations in the dynamics of exosome secretion, including their components, from these cells might underlie some of this risk. We found exosome size was not altered between common variant controls and R47Hhet variants, but the amount and constitution of exosomes secreted were different. Exosome quantities were rescued by incubation with an ATP donor or with lipids via a phosphatidylserine triggering receptor expressed on myeloid cells 2 ligand. Following a lipopolysaccharide or phagocytic cell stimulus, exosomes from common variant and R47Hhet microglia were found to contain cytokines, chemokines, APOE and triggering receptor expressed on myeloid cells 2. Differences were observed in the expression of CCL22, IL-1β and triggering receptor expressed on myeloid cells 2 between common variant and R47Hhet derived exosomes. Furthermore unlike common variant-derived exosomes, R47Hhet exosomes contained additional proteins linked to negative regulation of transcription and metabolic processes. Subsequent addition of exosomes to stressed neurones showed R47Hhet-derived exosomes to be less protective. These data have ramifications for the responses of microglia in Alzheimer's disease and may point to further targets for therapeutic intervention.
Collapse
Affiliation(s)
- Anna Mallach
- Department of Neuroinflammation, Queen Square Institute of Neurology, University College London, London WC1N 1PJ, UK
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, S-431 80, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, S-431 80, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80, Mölndal, Sweden
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 1PJ, UK
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 3BG, UK
- UK Dementia Research Institute University College London, London WC1E 6BT, UK
| | - John Hardy
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 1PJ, UK
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 3BG, UK
- UK Dementia Research Institute University College London, London WC1E 6BT, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
- NIHR University College London Hospitals Biomedical Research Centre, London, UK
- Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Thomas M Piers
- Department of Neuroinflammation, Queen Square Institute of Neurology, University College London, London WC1N 1PJ, UK
| | - Selina Wray
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 1PJ, UK
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Jennifer M Pocock
- Department of Neuroinflammation, Queen Square Institute of Neurology, University College London, London WC1N 1PJ, UK
| |
Collapse
|
24
|
Sánchez-Ceinos J, Rangel-Zuñiga OA, Clemente-Postigo M, Podadera-Herreros A, Camargo A, Alcalá-Diaz JF, Guzmán-Ruiz R, López-Miranda J, Malagón MM. miR-223-3p as a potential biomarker and player for adipose tissue dysfunction preceding type 2 diabetes onset. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 23:1035-1052. [PMID: 33614249 PMCID: PMC7868931 DOI: 10.1016/j.omtn.2021.01.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 01/14/2021] [Indexed: 12/20/2022]
Abstract
Circulating microRNAs (miRNAs) have been proposed as biomarkers for type 2 diabetes (T2D). Adipose tissue (AT), for which dysfunction is widely associated with T2D development, has been reported as a major source of circulating miRNAs. However, the role of dysfunctional AT in the altered pattern of circulating miRNAs associated with T2D onset remains unexplored. Herein, we investigated the relationship between T2D-associated circulating miRNAs and AT function, as well as the role of preadipocytes and adipocytes as secreting cells of candidate circulating miRNAs. Among the plasma miRNAs related to T2D onset in the CORonary Diet Intervention with Olive oil and cardiovascular PREVention (CORDIOPREV) cohort, baseline miR-223-3p levels (diminished in patients who next developed T2D [incident-T2D]) were significantly related to AT insulin resistance (IR). Baseline serum from incident-T2D participants induced inflammation and IR in 3T3-L1 adipocytes. We demonstrated that tumor necrosis factor (TNF)-α inhibited miR-223-3p secretion while enhancing miR-223-3p intracellular accumulation in 3T3-L1 (pre)adipocytes. Overexpression studies showed that an intracellular increase of miR-223-3p impaired glucose and lipid metabolism in these cells. Our findings provide mechanistic insights into the alteration of circulating miRNAs preceding T2D, unveiling both preadipocytes and adipocytes as miR-223-3p-secreting cells and suggesting that inflammation promotes miR-223-3p intracellular accumulation, which might contribute to (pre)adipocyte dysfunction and body metabolic dysregulation.
Collapse
Affiliation(s)
- Julia Sánchez-Ceinos
- Department of Cell Biology, Physiology, and Immunology; Maimónides Biomedical Research Institute of Córdoba (IMIBIC)/University of Córdoba/Reina Sofia University Hospital; Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN); Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Oriol A Rangel-Zuñiga
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN); Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain.,Lipids and Atherosclerosis Unit; Department of Internal Medicine, IMIBIC/Reina Sofia University Hospital/University of Córdoba; Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - Mercedes Clemente-Postigo
- Department of Cell Biology, Physiology, and Immunology; Maimónides Biomedical Research Institute of Córdoba (IMIBIC)/University of Córdoba/Reina Sofia University Hospital; Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain
| | - Alicia Podadera-Herreros
- Department of Cell Biology, Physiology, and Immunology; Maimónides Biomedical Research Institute of Córdoba (IMIBIC)/University of Córdoba/Reina Sofia University Hospital; Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN); Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Antonio Camargo
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN); Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain.,Lipids and Atherosclerosis Unit; Department of Internal Medicine, IMIBIC/Reina Sofia University Hospital/University of Córdoba; Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - Juan Francisco Alcalá-Diaz
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN); Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain.,Lipids and Atherosclerosis Unit; Department of Internal Medicine, IMIBIC/Reina Sofia University Hospital/University of Córdoba; Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - Rocío Guzmán-Ruiz
- Department of Cell Biology, Physiology, and Immunology; Maimónides Biomedical Research Institute of Córdoba (IMIBIC)/University of Córdoba/Reina Sofia University Hospital; Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN); Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - José López-Miranda
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN); Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain.,Lipids and Atherosclerosis Unit; Department of Internal Medicine, IMIBIC/Reina Sofia University Hospital/University of Córdoba; Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - María M Malagón
- Department of Cell Biology, Physiology, and Immunology; Maimónides Biomedical Research Institute of Córdoba (IMIBIC)/University of Córdoba/Reina Sofia University Hospital; Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN); Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| |
Collapse
|
25
|
Biadglegne F, König B, Rodloff AC, Dorhoi A, Sack U. Composition and Clinical Significance of Exosomes in Tuberculosis: A Systematic Literature Review. J Clin Med 2021; 10:E145. [PMID: 33406750 PMCID: PMC7795701 DOI: 10.3390/jcm10010145] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 12/21/2022] Open
Abstract
Tuberculosis (TB) remains a major health issue worldwide. In order to contain TB infections, improved vaccines as well as accurate and reliable diagnostic tools are desirable. Exosomes are employed for the diagnosis of various diseases. At present, research on exosomes in TB is still at the preliminary stage. Recent studies have described isolation and characterization of Mycobacterium tuberculosis (Mtb) derived exosomes in vivo and in vitro. Mtb-derived exosomes (Mtbexo) may be critical for TB pathogenesis by delivering mycobacterial-derived components to the recipient cells. Proteomic and transcriptomic analysis of Mtbexo have revealed a variety of proteins and miRNA, which are utilized by the TB bacteria for pathogenesis. Exosomes has been isolated in body fluids, are amenable for fast detection, and could contribute as diagnostic or prognostic biomarker to disease control. Extraction of exosomes from biological fluids is essential for the exosome research and requires careful standardization for TB. In this review, we summarized the different studies on Mtbexo molecules, including protein and miRNA and the method used to detect exosomes in biological fluids and cell culture supernatants. Thus, the detection of Mtbexo molecules in biological fluids may have a potential to expedite the diagnosis of TB infection. Moreover, the analysis of Mtbexo may generate new aspects in vaccine development.
Collapse
Affiliation(s)
- Fantahun Biadglegne
- College of Medicine and Health Sciences, Bahir Dar University, 79 Bahir Dar, Ethiopia
- Institute of Medical Microbiology and Epidemiology of Infectious Diseases, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (B.K.); (A.C.R.)
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany;
| | - Brigitte König
- Institute of Medical Microbiology and Epidemiology of Infectious Diseases, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (B.K.); (A.C.R.)
| | - Arne C. Rodloff
- Institute of Medical Microbiology and Epidemiology of Infectious Diseases, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (B.K.); (A.C.R.)
| | - Anca Dorhoi
- Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany;
| | - Ulrich Sack
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany;
| |
Collapse
|
26
|
Charest A. Experimental and Biological Insights from Proteomic Analyses of Extracellular Vesicle Cargos in Normalcy and Disease. ADVANCED BIOSYSTEMS 2020; 4:e2000069. [PMID: 32815324 PMCID: PMC8091982 DOI: 10.1002/adbi.202000069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 06/19/2020] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles (EVs) offer a vehicle for diagnostic and therapeutic utility. EVs carry bioactive cargo and an accrued interest in their characterization has emerged. Efforts at identifying EV-enriched protein or RNA led to a surprising realization that EVs are excessively heterogeneous in nature. This diversity is originally attributed to vesicle sizes but it is becoming evident that different classes of EVs vehiculate distinct molecular cargos. Therefore, one of the current challenges in EV research is their selective isolation in quantities sufficient for efficient downstream analyses. Many protocols have been developed; however, reproducibility between research groups can be difficult to reach and inter-studies analyses of data from different isolation protocols are unmanageable. Therefore, there is an unmet need to optimize and standardize methods and protocols for the isolation and purification of EVs. This review focuses on the diverse techniques and protocols used over the years to isolate and purify EVs with a special emphasis on their adequacy for proteomics applications. By combining recent advances in specific isolation methods that yield superior quality of EV preparations and mass spectrometry techniques, the field is now prepared for transformative advancements in establishing distinct categorization and cargo identification of subpopulations based on EV surface markers.
Collapse
|
27
|
Otahal A, Kramer K, Kuten-Pella O, Weiss R, Stotter C, Lacza Z, Weber V, Nehrer S, De Luna A. Characterization and Chondroprotective Effects of Extracellular Vesicles From Plasma- and Serum-Based Autologous Blood-Derived Products for Osteoarthritis Therapy. Front Bioeng Biotechnol 2020; 8:584050. [PMID: 33102466 PMCID: PMC7546339 DOI: 10.3389/fbioe.2020.584050] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022] Open
Abstract
Autologous blood products gain increasing interest in the field of regenerative medicine as well as in orthopedics, aesthetic surgery, and cosmetics. Currently, citrate-anticoagulated platelet-rich plasma (CPRP) preparations are often applied in osteoarthritis (OA), but more physiological and cell-free alternatives such as hyperacute serum (hypACT) are under development. Besides growth factors, blood products also bring along extracellular vesicles (EVs) packed with signal molecules, which open up a new level of complexity at evaluating the functional spectrum of blood products. Large proportions of EVs originated from platelets in CPRP and hypACT, whereas very low erythrocyte and monocyte-derived EVs were detected via flow cytometry. EV treatment of chondrocytes enhanced the expression of anabolic markers type II collagen, SRY-box transcription factor 9 (SOX9), and aggrecan compared to full blood products, but also the catabolic marker and tissue remodeling factor matrix metalloproteinase 3, whereas hypACT EVs prevented type I collagen expression. CPRP blood product increased SOX9 protein expression, in contrast to hypACT blood product. However, hypACT EVs induced SOX9 protein expression while preventing interleukin-6 secretion. The results indicate that blood EVs are sufficient to induce chondrogenic gene expression changes in OA chondrocytes, while preventing proinflammatory cytokine release compared to full blood product. This highlights the potential of autologous blood-derived EVs as regulators of cartilage extracellular matrix metabolism and inflammation, as well as candidates for new cell-free therapeutic approaches for OA.
Collapse
Affiliation(s)
- Alexander Otahal
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria
| | - Karina Kramer
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria
| | - Olga Kuten-Pella
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria.,OrthoSera GmbH, Krems an der Donau, Austria
| | - René Weiss
- Center for Biomedical Technology, Department for Biomedical Research, Danube University Krems, Krems an der Donau, Austria
| | - Christoph Stotter
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria
| | - Zsombor Lacza
- Deptartment Sports Physiology, University of Physical Education, Budapest, Hungary
| | - Viktoria Weber
- Center for Biomedical Technology, Department for Biomedical Research, Danube University Krems, Krems an der Donau, Austria
| | - Stefan Nehrer
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria
| | - Andrea De Luna
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria
| |
Collapse
|
28
|
Elashiry M, Elashiry MM, Elsayed R, Rajendran M, Auersvald C, Zeitoun R, Rashid MH, Ara R, Meghil MM, Liu Y, Arbab AS, Arce RM, Hamrick M, Elsalanty M, Brendan M, Pacholczyk R, Cutler CW. Dendritic cell derived exosomes loaded with immunoregulatory cargo reprogram local immune responses and inhibit degenerative bone disease in vivo. J Extracell Vesicles 2020; 9:1795362. [PMID: 32944183 PMCID: PMC7480413 DOI: 10.1080/20013078.2020.1795362] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Chronic bone degenerative diseases represent a major threat to the health and well-being of the population, particularly those with advanced age. This study isolated exosomes (EXO), natural nano-particles, from dendritic cells, the “directors” of the immune response, to examine the immunobiology of DC EXO in mice, and their ability to reprogram immune cells responsible for experimental alveolar bone loss in vivo. Distinct DC EXO subtypes including immune-regulatory (regDC EXO), loaded with TGFB1 and IL10 after purification, along with immune stimulatory (stimDC EXO) and immune “null” immature (iDCs EXO) unmodified after purification, were delivered via I.V. route or locally into the soft tissues overlying the alveolar bone. Locally administrated regDC EXO showed high affinity for inflamed sites, and were taken up by both DCs and T cells in situ. RegDC EXO-encapsulated immunoregulatory cargo (TGFB1 and IL10) was protected from proteolytic degradation. Moreover, maturation of recipient DCs and induction of Th17 effectors was suppressed by regDC EXO, while T-regulatory cell recruitment was promoted, resulting in inhibition of bone resorptive cytokines and reduction in osteoclastic bone loss. This work is the first demonstration of DC exosome-based therapy for a degenerative alveolar bone disease and provides the basis for a novel treatment strategy.
Collapse
Affiliation(s)
- Mahmoud Elashiry
- Department of Periodontics, Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia at Augusta University, Augusta, GA, USA
| | - Mohamed M Elashiry
- Department of Periodontics, Dental College of Georgia at Augusta University, GA, USA, Department of Endodontics, Faculty of Dentistry, Ain Shams University, Cairo, Egypt
| | - Ranya Elsayed
- Department of Periodontics, Dental College of Georgia at Augusta University, GA, USA
| | - Mythily Rajendran
- Department of Periodontics, Dental College of Georgia at Augusta University, GA, USA
| | - Carol Auersvald
- Department of Periodontics, Dental College of Georgia at Augusta University, GA, USA
| | - Rana Zeitoun
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia at Augusta University, Department of Fixed Prosthodontics, Faculty of Dentistry, Ain Shams University, Cairo, Egypt
| | - Mohammad H Rashid
- Department of Biochemistry & Molecular Biology, Georgia Cancer Center, Augusta, GA, USA
| | - Roxan Ara
- Department of Biochemistry & Molecular Biology, Georgia Cancer Center, Augusta, GA, USA
| | - Mohamed M Meghil
- Department of Periodontics, Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia at Augusta University, Augusta, GA, USA
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, GA, USA
| | - Ali S Arbab
- Department of Biochemistry & Molecular Biology, Georgia Cancer Center, Augusta, GA, USA
| | - Roger M Arce
- Department of Periodontics and Oral Hygiene, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Mark Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, GA, USA
| | - Mohammed Elsalanty
- Department of Periodontics, Dental College of Georgia at Augusta University, GA, USA
| | - Marshall Brendan
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, GA, USA
| | - Rafal Pacholczyk
- Georgia Cancer Center, Augusta, GA, USA.,Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia at Augusta University, GA, USA
| | - Christopher W Cutler
- Department of Periodontics, Dental College of Georgia at Augusta University, GA, USA
| |
Collapse
|
29
|
Rodovalho VDR, da Luz BSR, Rabah H, do Carmo FLR, Folador EL, Nicolas A, Jardin J, Briard-Bion V, Blottière H, Lapaque N, Jan G, Le Loir Y, de Carvalho Azevedo VA, Guédon E. Extracellular Vesicles Produced by the Probiotic Propionibacterium freudenreichii CIRM-BIA 129 Mitigate Inflammation by Modulating the NF-κB Pathway. Front Microbiol 2020; 11:1544. [PMID: 32733422 PMCID: PMC7359729 DOI: 10.3389/fmicb.2020.01544] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/15/2020] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs) are nanometric spherical structures involved in intercellular communication, whose production is considered to be a widespread phenomenon in living organisms. Bacterial EVs are associated with several processes that include survival, competition, pathogenesis, and immunomodulation. Among probiotic Gram-positive bacteria, some Propionibacterium freudenreichii strains exhibit anti-inflammatory activity, notably via surface proteins such as the surface-layer protein B (SlpB). We have hypothesized that, in addition to surface exposure and secretion of proteins, P. freudenreichii may produce EVs and thus export immunomodulatory proteins to interact with the host. In order to demonstrate their production in this species, EVs were purified from cell-free culture supernatants of the probiotic strain P. freudenreichii CIRM-BIA 129, and their physicochemical characterization, using transmission electron microscopy and nanoparticle tracking analysis (NTA), revealed shapes and sizes typical of EVs. Proteomic characterization showed that EVs contain a broad range of proteins, including immunomodulatory proteins such as SlpB. In silico protein-protein interaction predictions indicated that EV proteins could interact with host proteins, including the immunomodulatory transcription factor NF-κB. This potential interaction has a functional significance because EVs modulate inflammatory responses, as shown by IL-8 release and NF-κB activity, in HT-29 human intestinal epithelial cells. Indeed, EVs displayed an anti-inflammatory effect by modulating the NF-κB pathway; this was dependent on their concentration and on the proinflammatory inducer (LPS-specific). Moreover, while this anti-inflammatory effect partly depended on SlpB, it was not abolished by EV surface proteolysis, suggesting possible intracellular sites of action for EVs. This is the first report on identification of P. freudenreichii-derived EVs, alongside their physicochemical, biochemical and functional characterization. This study has enhanced our understanding of the mechanisms associated with the probiotic activity of P. freudenreichii and identified opportunities to employ bacterial-derived EVs for the development of bioactive products with therapeutic effects.
Collapse
Affiliation(s)
- Vinícius de Rezende Rodovalho
- INRAE, Institut Agro, STLO, Rennes, France.,Laboratory of Cellular and Molecular Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Brenda Silva Rosa da Luz
- INRAE, Institut Agro, STLO, Rennes, France.,Laboratory of Cellular and Molecular Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Fillipe Luiz Rosa do Carmo
- Laboratory of Cellular and Molecular Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Edson Luiz Folador
- Biotechnology Center, Federal University of Paraíba, João Pessoa, Brazil
| | | | | | | | - Hervé Blottière
- INRAE, AgroParisTech, Paris-Saclay University, Micalis Institute, Jouy-en-Josas, France
| | - Nicolas Lapaque
- INRAE, AgroParisTech, Paris-Saclay University, Micalis Institute, Jouy-en-Josas, France
| | | | | | - Vasco Ariston de Carvalho Azevedo
- Laboratory of Cellular and Molecular Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | |
Collapse
|
30
|
Xu L, Shoaie N, Jahanpeyma F, Zhao J, Azimzadeh M, Al Jamal KT. Optical, electrochemical and electrical (nano)biosensors for detection of exosomes: A comprehensive overview. Biosens Bioelectron 2020; 161:112222. [PMID: 32365010 DOI: 10.1016/j.bios.2020.112222] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/13/2022]
Abstract
Exosomes are small extracellular vesicles involved in many physiological activities of cells in the human body. Exosomes from cancer cells have great potential to be applied in clinical diagnosis, early cancer detection and target identification for molecular therapy. While this field is gaining increasing interests from both academia and industry, barriers such as supersensitive detection techniques and highly-efficient isolation methods remain. In the clinical settings, there is an urgent need for rapid analysis, reliable detection and point-of-care testing (POCT). With these challenges to be addressed, this article aims to review recent developments and technical breakthroughs including optical, electrochemical and electrical biosensors for exosomes detection in the field of cancer and other diseases and demonstrate how nanobiosensors could enhance the performance of conventional sensors. Working strategies, limit of detections, advantages and shortcomings of the studies are summarized. New trends, challenges and future perspectives of exosome-driven POCT in liquid biopsy have been discussed.
Collapse
Affiliation(s)
- Lizhou Xu
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, United Kingdom
| | - Nahid Shoaie
- Department of Biotechnology, Tarbiat Modares University of Medical Science, Tehran, Iran
| | - Fatemeh Jahanpeyma
- Department of Biotechnology, Tarbiat Modares University of Medical Science, Tehran, Iran
| | - Junjie Zhao
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, United Kingdom
| | - Mostafa Azimzadeh
- Medical Nanotechnology & Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, 89195-999, Yazd, Iran; Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, 89195-999, Yazd, Iran; Department of Advanced Medical Sciences and Technologies, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, 8916188635, Yazd, Iran.
| | - Khuloud T Al Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, United Kingdom.
| |
Collapse
|
31
|
The Course of Circulating Small Extracellular Vesicles in Patients Undergoing Surgical Aortic Valve Replacement. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6381396. [PMID: 32382562 PMCID: PMC7193280 DOI: 10.1155/2020/6381396] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 02/12/2020] [Accepted: 02/19/2020] [Indexed: 01/05/2023]
Abstract
In the last years, increasing efforts have been devoted to investigating the role of small extracellular vesicles (sEVs) in cardiovascular diseases. These nano-sized particles (30-150 nm), secreted by different cell types, contain signalling molecules that enable participation in intercellular communication processes. In this study, we examined the course of circulating sEVs in patients undergoing surgical aortic valve replacement (SAVR) and correlated them with echocardiographic and standard blood parameters. Peripheral blood samples were collected from 135 patients undergoing SAVR preoperatively and at three follow-up points. Circulating sEVs were precipitated using Exoquick™ exosome isolation reagent and analyzed by nanoparticle tracking analysis (NTA). Our findings indicate that no more than 7 days after SAVR, there was a marked increase of circulating sEVs before returning to initial values after 3 months. Further, shear stress is not a trigger for the formation and release of circulating sEVs. Moreover, we pointed out a correlation between circulating sEVs and erythrocytes as well as LDH and creatinine levels in peripheral blood. Finally, all patients with a moderate prosthesis-patient mismatch as well as with an impaired left ventricular mass regression had lower levels of circulating sEVs 3 months after SAVR compared to their respective status before surgery. We conclude that in patients with aortic valve stenosis (AVS), sEVs may play an important part in mediating cell-cell communication and SAVR may have a crucial and lasting impact on their circulating levels. Besides, lower levels of sEVs portend to be associated with inferior recovery after major surgical interventions. The additional use of circulating sEVs beyond echocardiographic and laboratory parameters could have a prognostic value to estimate adverse outcomes in patients undergoing SAVR.
Collapse
|
32
|
Vucetic A, Filho ADSL, Dong G, Olivier M. Isolation of Extracellular Vesicles from Leishmania spp. Methods Mol Biol 2020; 2116:555-574. [PMID: 32221942 DOI: 10.1007/978-1-0716-0294-2_33] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Exosomes, a class of extracellular vesicles, are released by eukaryotes, bacteria, and archaea, as evident from both in vitro and in vivo studies. These nano-sized double-membraned vesicles play an important role in cell-to-cell communication, dysregulation of the immune system, and pathogenesis in a number of diseases, including leishmaniasis. Leishmania is a genus of obligate intracellular parasites, which infect host macrophages, are transmitted through the bite of a sandfly, and are shown to secrete exosomes with immunomodulatory activities. Given the importance of these vesicles in Leishmania spp. virulence, it is necessary to perform appropriate isolation and characterization in order to further study their relevance in the parasite's infectious life cycle. In this chapter, we describe four methods for the isolation of extracellular vesicles derived from Leishmania species including ultracentrifugation, polyethylene glycol-based precipitation, size-exclusion chromatography, and sucrose-gradient fractionation. Further, we describe the preparation of isolated samples for characterization by nanoparticle tracking analysis, transmission electron microscopy, and proteomic profiling.
Collapse
Affiliation(s)
- Andrea Vucetic
- Infectious Diseases and Immunity in Global Health (IDIGH) Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Alonso Da Silva Lira Filho
- Infectious Diseases and Immunity in Global Health (IDIGH) Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - George Dong
- Infectious Diseases and Immunity in Global Health (IDIGH) Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Martin Olivier
- Infectious Diseases and Immunity in Global Health (IDIGH) Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
33
|
Good ME, Musante L, Salvia SL, Howell NL, Carey RM, Le TH, Isakson BE, Erdbrügger U. Circulating Extracellular Vesicles in Normotension Restrain Vasodilation in Resistance Arteries. Hypertension 2020; 75:218-228. [PMID: 31760883 PMCID: PMC7158164 DOI: 10.1161/hypertensionaha.119.13363] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 10/19/2019] [Indexed: 12/26/2022]
Abstract
Extracellular vesicles (EVs) have been described as novel biomarkers and bioactivators in vascular dysfunction in hypertension. However, the mechanism(s) by which EVs affect vascular function is unknown. To examine the effects of EVs on endothelial-dependent vasodilation (acetylcholine), we isolated circulating EVs from platelet-poor plasma using a low centrifugation speed (17 000g) and mesenteric resistance arteries from 12-week-old normotensive WKYs (Wistar-Kyoto rats) and SHRs (spontaneously hypertensive rats). Arteries were cannulated on a pressure myograph, and EVs were added to the vessel lumen and circulating bath. We found that circulating EVs from normotensive WKY reduced vasodilation of normotensive WKY arteries but had no effect on hypertensive SHR arteries. In contrast, EVs from hypertensive SHR failed to reduce vasodilation of arteries from both WKY and SHR. The restraining effect on vasodilation by EVs from normotensive WKY may be mediated by inhibition of eNOS (endothelial NO synthase), as addition of L-nitro-arginine methyl ester did not provide any additive effect. Moreover, circulating EVs from normotensive 6-week-old SHR-an age where SHRs have not yet developed hypertension-had similar restraining effect on vasodilation. In addition, delipidation of EVs did not alter the restraining effect of EVs from WKY but did restore the restraining effect of EVs from SHR. Finally, EVs from normotensive humans also restrained vasodilation of normotensive mouse arteries-an effect not observed in EVs from hypertensive humans. Taken together, our data support a vasoactive role of EVs that is altered in hypertension.
Collapse
Affiliation(s)
- Miranda E Good
- Robert M Berne Cardiovascular Research Center, University of Virginia School of Medicine
| | - Luca Musante
- Department of Medicine, Division of Nephrology, University of Virginia School of Medicine
| | - Sabrina La Salvia
- Department of Medicine, Division of Nephrology, University of Virginia School of Medicine
| | - Nancy L Howell
- Department of Medicine, Division of Endocrinology and Metabolism, University of Virginia School of Medicine
| | - Robert M Carey
- Department of Medicine, Division of Endocrinology and Metabolism, University of Virginia School of Medicine
| | - Thu H Le
- Department of Medicine, Division of Nephrology, University of Rochester, New York
| | - Brant E Isakson
- Robert M Berne Cardiovascular Research Center, University of Virginia School of Medicine
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine
| | - Uta Erdbrügger
- Department of Medicine, Division of Nephrology, University of Virginia School of Medicine
| |
Collapse
|
34
|
Lin F, Yin HB, Li XY, Zhu GM, He WY, Gou X. Bladder cancer cell‑secreted exosomal miR‑21 activates the PI3K/AKT pathway in macrophages to promote cancer progression. Int J Oncol 2019; 56:151-164. [PMID: 31814034 PMCID: PMC6910194 DOI: 10.3892/ijo.2019.4933] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 11/08/2019] [Indexed: 12/11/2022] Open
Abstract
Tumour-associated macrophages (TAMs) compose a major component of the tumour microenvironment and form in this microenvironment prior to cancer metastasis. However, the detailed mechanisms of TAM remodelling in the context of bladder cancer have not been clearly defined. The present study collected exosomes from the conditioned medium of human bladder T24 cancer cells. The effects of macrophages treated with exosomes derived from T24 cells on bladder cancer cell migration and invasion were analysed by Transwell assays. The expression levels of endogenous and exosomal microRNA-21 (miR-21) were examined by reverse transcription-quantitative PCR, while the expression level of the target protein was analysed by western blot analysis. Luciferase reporter plasmids and mutants were used to confirm direct targeting. The effects of miR-21 on bladder cancer cell migration and invasion were analysed by Transwell and Matrigel assays following miR-21 transfection. It was identified that exosomes derived from bladder cancer cells polarized THP-1 cell-derived macrophages into the M2 phenotype, and TAM-mediated pro-migratory and pro-invasive activity was determined. Moreover, it was found that miR-21 was highly expressed in exosomes derived from bladder cancer cells as well as in macrophages treated with exosomes. In addition, macrophages transfected with miR-21 exhibited M2 polarization and promoted T24 cell migratory and invasive ability. Mechanistically, exosomal miR-21 derived from bladder cancer cells inhibited phosphatase and tensin homolog activation of the PI3K/AKT signalling pathway in macrophages and enhanced STAT3 expression to promote M2 phenotypic polarization. The present results suggest that exosomal miR-21 can promote cancer progression by polarizing TAMs.
Collapse
Affiliation(s)
- Fan Lin
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hu-Bin Yin
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xin-Yuan Li
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Gong-Min Zhu
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Wei-Yang He
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xin Gou
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
35
|
Yao B, Qu S, Hu R, Gao W, Jin S, Liu M, Zhao Q. A panel of miRNAs derived from plasma extracellular vesicles as novel diagnostic biomarkers of lung adenocarcinoma. FEBS Open Bio 2019; 9:2149-2158. [PMID: 31677346 PMCID: PMC6886307 DOI: 10.1002/2211-5463.12753] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 10/31/2019] [Indexed: 12/13/2022] Open
Abstract
Lung cancer is the leading cause of cancer‐related morbidity and mortality worldwide, with lung adenocarcinoma (LUAD) being the most common histological subtype (approximately 40%). In the absence of reliable screening biomarkers for early diagnosis, most patients with LUAD are inevitably diagnosed at an advanced stage. MicroRNAs (miRNAs) encapsulated within plasma‐derived extracellular vesicles (EVs) may be suitable for use as noninvasive diagnostic biomarkers for aggressive malignancies, including LUAD. In this study, we first investigated the miRNA profiles of plasma‐derived EVs from LUAD patients and healthy donors, and then systematically evaluated the expression patterns of selected plasma‐derived EV miRNAs in a large cohort of patients with LUAD and healthy controls. Notably, we observed that miR‐451a, miR‐194‐5p, and miR‐486‐5p were significantly increased in EVs from LUAD patients, compared to healthy controls. The area under the curve values for the three miRNAs were 0.9040 (95% confidence interval [CI], 0.8633–0.9447) for miR‐451a, 0.7492 (95% CI, 0.6992–0.7992) for miR‐194‐5p, and 0.9574 (95% CI, 0.9378–0.9769) for miR‐486‐5p, while the AUC of the combination of these three miRNAs was 0.9650. Thus, these results suggest that these EV miRNAs may be promising candidates for the development of highly effective, noninvasive biomarkers for early LUAD diagnosis. Lung cancer is the leading cause of cancer‐related morbidity and mortality worldwide, with lung adenocarcinoma (LUAD) being the most common histological subtype (approximately 40%). In this study, we observed that miR‐451a, miR‐194‐5p, and miR‐486‐5p in extracellular vesicles derived from plasma may be suitable as highly effective and noninvasive biomarkers for early LUAD diagnosis.![]()
Collapse
Affiliation(s)
- Bing Yao
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, China
| | - Shuang Qu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, China
| | - Ruifeng Hu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, China
| | - Wen Gao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, China
| | - Shidai Jin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, China
| | - Ming Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, China
| | - Quan Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, China
| |
Collapse
|
36
|
Gómez-de-Mariscal E, Maška M, Kotrbová A, Pospíchalová V, Matula P, Muñoz-Barrutia A. Deep-Learning-Based Segmentation of Small Extracellular Vesicles in Transmission Electron Microscopy Images. Sci Rep 2019; 9:13211. [PMID: 31519998 PMCID: PMC6744556 DOI: 10.1038/s41598-019-49431-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 08/05/2019] [Indexed: 02/07/2023] Open
Abstract
Small extracellular vesicles (sEVs) are cell-derived vesicles of nanoscale size (~30-200 nm) that function as conveyors of information between cells, reflecting the cell of their origin and its physiological condition in their content. Valuable information on the shape and even on the composition of individual sEVs can be recorded using transmission electron microscopy (TEM). Unfortunately, sample preparation for TEM image acquisition is a complex procedure, which often leads to noisy images and renders automatic quantification of sEVs an extremely difficult task. We present a completely deep-learning-based pipeline for the segmentation of sEVs in TEM images. Our method applies a residual convolutional neural network to obtain fine masks and use the Radon transform for splitting clustered sEVs. Using three manually annotated datasets that cover a natural variability typical for sEV studies, we show that the proposed method outperforms two different state-of-the-art approaches in terms of detection and segmentation performance. Furthermore, the diameter and roundness of the segmented vesicles are estimated with an error of less than 10%, which supports the high potential of our method in biological applications.
Collapse
Affiliation(s)
- Estibaliz Gómez-de-Mariscal
- Bioengineering and Aerospace Engineering Department, Universidad Carlos III de Madrid, Leganés, 28911, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, 28007, Spain
| | - Martin Maška
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, 602 00, Czech Republic
| | - Anna Kotrbová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 611 37, Czech Republic
| | - Vendula Pospíchalová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 611 37, Czech Republic
| | - Pavel Matula
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, 602 00, Czech Republic
| | - Arrate Muñoz-Barrutia
- Bioengineering and Aerospace Engineering Department, Universidad Carlos III de Madrid, Leganés, 28911, Spain.
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, 28007, Spain.
| |
Collapse
|
37
|
Bai Y, Lu Y, Wang K, Cheng Z, Qu Y, Qiu S, Zhou L, Wu Z, Liu H, Zhao J, Mao H. Rapid Isolation and Multiplexed Detection of Exosome Tumor Markers Via Queued Beads Combined with Quantum Dots in a Microarray. NANO-MICRO LETTERS 2019; 11:59. [PMID: 34137993 PMCID: PMC7770845 DOI: 10.1007/s40820-019-0285-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 06/10/2019] [Indexed: 05/04/2023]
Abstract
Tumor-derived exosomes are actively involved in cancer progression and metastasis and have emerged as a promising marker for cancer diagnosis in liquid biopsy. Because of their nanoscale size, complex biogenesis, and methodological limitations related to exosome isolation and detection, advancements in their analysis remain slow. Microfluidic technology offers a better analytic approach compared with conventional methods. Here, we developed a bead-based microarray for exosome isolation and multiplexed tumor marker detection. Using this method, exosomes are isolated by binding to antibodies on the bead surface, and tumor markers on the exosomes are detected through quantum dot (QD) probes. The beads are then uniformly trapped and queued among micropillars in the chip. This design benefits fluorescence observation by dispersing the signals into every single bead, thereby avoiding optical interference and enabling more accurate test results. We analyzed exosomes in the cell culture supernatant of lung cancer and endothelial cell lines, and different lung cancer markers labeled with three QD probes were used to conduct multiplexed detection of exosome surface protein markers. Lung cancer-derived samples showed much higher (~ sixfold-tenfold) fluorescence intensity than endothelial cell samples, and different types of lung cancer samples showed distinctive marker expression levels. Additionally, using the chip to detect clinical plasma samples from cancer patients showed good diagnostic power and revealed a well consistency with conventional tests for serological markers. These results provide insight into a promising method for exosome tumor marker detection and early-stage cancer diagnosis.
Collapse
Affiliation(s)
- Yanan Bai
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Yunxing Lu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Kun Wang
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Zule Cheng
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Youlan Qu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China
- School of Stomatology, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Shihui Qiu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China
| | - Lin Zhou
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China
| | - Zhenhua Wu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China
| | - Huiying Liu
- School of Stomatology, Dalian Medical University, Dalian, 116044, People's Republic of China.
| | - Jianlong Zhao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China.
| | - Hongju Mao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China.
| |
Collapse
|
38
|
Septin 9 isoforms promote tumorigenesis in mammary epithelial cells by increasing migration and ECM degradation through metalloproteinase secretion at focal adhesions. Oncogene 2019; 38:5839-5859. [PMID: 31285548 PMCID: PMC6859949 DOI: 10.1038/s41388-019-0844-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 02/07/2023]
Abstract
The cytoskeletal interacting protein Septin 9 (SEPT9), a member of the septin gene family, has been proposed to have oncogenic functions. It is a known hot spot of retroviral tagging insertion and a fusion partner of both de novo and therapy-induced mixed lineage leukemia (MLL). Of all septins, SEPT9 holds the strongest link to cancer, especially breast cancer. Murine models of breast cancer frequently exhibit Sept9 amplification in the form of double minute chromosomes, and about 20% of human breast cancer display genomic amplification and protein over expression at the SEPT9 locus. Yet, a clear mechanism by which SEPT9 elicits tumor-promoting functions is lacking. To obtain unbiased insights on molecular signatures of SEPT9 upregulation in breast tumors, we overexpressed several of its isoforms in breast cancer cell lines. Global transcriptomic profiling supports a role of SEPT9 in invasion. Functional studies reveal that SEPT9 upregulation is sufficient to increase degradation of the extracellular matrix, while SEPT9 downregulation inhibits this process. The degradation pattern is peripheral and associated with focal adhesions (FA), where it is coupled with increased expression of matrix metalloproteinases. SEPT9 overexpression induces MMP upregulation in human tumors and in culture models and promotes MMP3 secretion to the media at FAs. Downregulation of SEPT9 or chemical inhibition of septin filament assembly impairs recruitment of MMP3 to FAs. Our results indicate that SEPT9 promotes upregulation and both trafficking and secretion of MMPs near FAs, thus enhancing migration and invasion of breast cancer cells.
Collapse
|
39
|
Vibrio cholerae derived outer membrane vesicles modulate the inflammatory response of human intestinal epithelial cells by inducing microRNA-146a. Sci Rep 2019; 9:7212. [PMID: 31076615 PMCID: PMC6510749 DOI: 10.1038/s41598-019-43691-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 04/23/2019] [Indexed: 01/15/2023] Open
Abstract
The small intestinal epithelium of Vibrio cholerae infected patients expresses the immunomodulatory microRNAs miR-146a and miR-155 at acute stage of disease. V. cholerae release outer membrane vesicles (OMVs) that serve as vehicles for translocation of virulence factors including V. cholerae cytolysin (VCC). The aim was to investigate whether OMVs, with and/or without VCC-cargo could be responsible for induction of microRNAs in intestinal epithelial cells and thereby contribute to immunomodulation. Polarized tight monolayers of T84 cells were challenged with OMVs of wildtype and a VCC deletion mutant of the non-O1/non-O139 (NOVC) V. cholerae strain V:5/04 and with soluble VCC. OMVs, with and without VCC-cargo, caused significantly increased levels of miR-146a. Increase was seen already after 2 hours challenge with OMVs and persisted after 12 hours. Challenge with soluble VCC caused significant increases in interleukin-8 (IL-8), tumour necrosis factor-α (TNF-α), CCL20, IL-1β, and IRAK2 mRNA levels while challenge with OMVs did not cause increases in expression levels of any of these mRNAs. These results suggest that V. cholerae bacteria release OMVs that induce miR-146a in order to pave the way for colonization by reducing the strength of an epithelial innate immune defence reaction and also preventing inflammation in the mucosa that factors like VCC can evoke.
Collapse
|
40
|
Parry HA, Mobley CB, Mumford PW, Romero MA, Haun CT, Zhang Y, Roberson PA, Zempleni J, Ferrando AA, Vechetti IJ, McCarthy JJ, Young KC, Roberts MD, Kavazis AN. Bovine Milk Extracellular Vesicles (EVs) Modification Elicits Skeletal Muscle Growth in Rats. Front Physiol 2019; 10:436. [PMID: 31040795 PMCID: PMC6476979 DOI: 10.3389/fphys.2019.00436] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 03/29/2019] [Indexed: 12/12/2022] Open
Abstract
The current study investigated how bovine milk extracellular vesicles (EVs) affected rotarod performance and biomarkers of skeletal muscle physiology in young, growing rats. Twenty-eight-day Fisher 344 rats were provided an AIN-93G-based diet for 4 weeks that either remained unadulterated [EVs and RNA-sufficient (ERS; n = 12)] or was sonicated [EVs and RNA-depleted (ERD; n = 12)]. Prior to (PRE) and on the last day of the intervention (POST), animals were tested for maximal rotarod performance. Following the feeding period, the gastrocnemius muscle was analyzed at the histological, biochemical, and molecular levels and was also used to measure mitochondrial function and reactive oxygen species (ROS) emission. A main effect of time was observed for rotarod time (PRE > POST, p = 0.001). Terminal gastrocnemius mass was unaffected by diet, although gastrocnemius muscle fiber cross sectional area was 11% greater (p = 0.018) and total RNA (a surrogate of ribosome density) was 24% greater (p = 0.001) in ERD. Transcriptomic analysis of the gastrocnemius indicated that 22 mRNAs were significantly greater in ERS versus ERD (p < 0.01), whereas 55 mRNAs were greater in ERD versus ERS (p < 0.01). There were no differences in gastrocnemius citrate synthase activity or mitochondrial coupling (respiratory control ratio), although mitochondrial ROS production was lower in ERD gastrocnemius (p = 0.016), which may be explained by an increase in glutathione peroxidase protein levels (p = 0.020) in ERD gastrocnemius. Dietary EVs profiling confirmed that sonication in the ERD diet reduced EVs content by ∼60%. Our findings demonstrate that bovine milk EVs depletion through sonication elicits anabolic and transcriptomic effects in the gastrocnemius muscle of rapidly maturing rats. While this did not translate into a functional outcome between diets (i.e., rotarod performance), longer feeding periods may be needed to observe such functional effects.
Collapse
Affiliation(s)
- Hailey A. Parry
- School of Kinesiology, Auburn University, Auburn, AL, United States
| | - C. Brooks Mobley
- School of Kinesiology, Auburn University, Auburn, AL, United States
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Petey W. Mumford
- School of Kinesiology, Auburn University, Auburn, AL, United States
| | | | - Cody T. Haun
- School of Kinesiology, Auburn University, Auburn, AL, United States
| | - Yufeng Zhang
- School of Kinesiology, Auburn University, Auburn, AL, United States
| | - Paul A. Roberson
- School of Kinesiology, Auburn University, Auburn, AL, United States
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska–Lincoln, Lincoln, NE, United States
| | - Arny A. Ferrando
- Donald W. Reynolds Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AK, United States
| | - Ivan J. Vechetti
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, United States
| | - John J. McCarthy
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Kaelin C. Young
- School of Kinesiology, Auburn University, Auburn, AL, United States
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine-Auburn Campus, Auburn, AL, United States
| | - Michael D. Roberts
- School of Kinesiology, Auburn University, Auburn, AL, United States
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine-Auburn Campus, Auburn, AL, United States
| | - Andreas N. Kavazis
- School of Kinesiology, Auburn University, Auburn, AL, United States
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine-Auburn Campus, Auburn, AL, United States
| |
Collapse
|
41
|
Bachurski D, Schuldner M, Nguyen PH, Malz A, Reiners KS, Grenzi PC, Babatz F, Schauss AC, Hansen HP, Hallek M, Pogge von Strandmann E. Extracellular vesicle measurements with nanoparticle tracking analysis - An accuracy and repeatability comparison between NanoSight NS300 and ZetaView. J Extracell Vesicles 2019; 8:1596016. [PMID: 30988894 PMCID: PMC6450530 DOI: 10.1080/20013078.2019.1596016] [Citation(s) in RCA: 307] [Impact Index Per Article: 61.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 02/14/2019] [Accepted: 03/03/2019] [Indexed: 12/13/2022] Open
Abstract
The expanding field of extracellular vesicle (EV) research needs reproducible and accurate methods to characterize single EVs. Nanoparticle Tracking Analysis (NTA) is commonly used to determine EV concentration and diameter. As the EV field is lacking methods to easily confirm and validate NTA data, questioning the reliability of measurements remains highly important. In this regard, a comparison addressing measurement quality between different NTA devices such as Malvern’s NanoSight NS300 or Particle Metrix’ ZetaView has not yet been conducted. To evaluate the accuracy and repeatability of size and concentration determinations of both devices, we employed comparative methods including transmission electron microscopy (TEM) and single particle interferometric reflectance imaging sensing (SP-IRIS) by ExoView. Multiple test measurements with nanospheres, liposomes and ultracentrifuged EVs from human serum and cell culture supernatant were performed. Additionally, serial dilutions and freeze-thaw cycle-dependent EV decrease were measured to determine the robustness of each system. Strikingly, NanoSight NS300 exhibited a 2.0–2.1-fold overestimation of polystyrene and silica nanosphere concentration. By measuring serial dilutions of EV samples, we demonstrated higher accuracy in concentration determination by ZetaView (% BIAS range: 2.7–8.5) in comparison with NanoSight NS300 (% BIAS range: 32.9–36.8). The concentration measurements by ZetaView were also more precise (% CV range: 0.0–4.7) than measurements by NanoSight NS300 (% CV range: 5.4–10.7). On the contrary, quantitative TEM imaging indicated more accurate EV sizing by NanoSight NS300 (% DTEM range: 79.5–134.3) compared to ZetaView (% DTEM range: 111.8–205.7), while being equally repeatable (NanoSight NS300% CV range: 0.8–6.7; ZetaView: 1.4–7.8). However, both devices failed to report a peak EV diameter below 60 nm compared to TEM and SP-IRIS. Taken together, NTA devices differ strongly in their hardware and software affecting measuring results. ZetaView provided a more accurate and repeatable depiction of EV concentration, whereas NanoSight NS300 supplied size measurements of higher resolution.
Collapse
Affiliation(s)
- Daniel Bachurski
- Department I of Internal Medicine, University Hospital of Cologne, Center for Integrated Oncology Cologne-Bonn, CECAD Center of Excellence on ''Cellular Stress Responses in Aging-Associated Diseases'', Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Maximiliane Schuldner
- Department I of Internal Medicine, University of Cologne, Cologne, Germany.,Experimental Tumor Research, Center for Tumor Biology and Immunology, Department of Hematology, Oncology and Immunology, Philipps University Marburg, Marburg, Germany
| | - Phuong-Hien Nguyen
- Department I of Internal Medicine, University Hospital of Cologne, Center for Integrated Oncology Cologne-Bonn, CECAD Center of Excellence on ''Cellular Stress Responses in Aging-Associated Diseases'', Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Alexandra Malz
- Experimental Tumor Research, Center for Tumor Biology and Immunology, Department of Hematology, Oncology and Immunology, Philipps University Marburg, Marburg, Germany
| | - Katrin S Reiners
- Department I of Internal Medicine, University of Cologne, Cologne, Germany.,Institute for Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| | - Patricia C Grenzi
- Department I of Internal Medicine, University of Cologne, Cologne, Germany
| | - Felix Babatz
- CECAD Center of Excellence on ''Cellular Stress Responses in Aging-Associated Diseases'', University of Cologne, Cologne, Germany
| | - Astrid C Schauss
- CECAD Center of Excellence on ''Cellular Stress Responses in Aging-Associated Diseases'', University of Cologne, Cologne, Germany
| | - Hinrich P Hansen
- Department I of Internal Medicine, University Hospital of Cologne, Center for Integrated Oncology Cologne-Bonn, CECAD Center of Excellence on ''Cellular Stress Responses in Aging-Associated Diseases'', Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Michael Hallek
- Department I of Internal Medicine, University Hospital of Cologne, Center for Integrated Oncology Cologne-Bonn, CECAD Center of Excellence on ''Cellular Stress Responses in Aging-Associated Diseases'', Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Elke Pogge von Strandmann
- Experimental Tumor Research, Center for Tumor Biology and Immunology, Department of Hematology, Oncology and Immunology, Philipps University Marburg, Marburg, Germany
| |
Collapse
|
42
|
Yang Y, Zhang L, La X, Li Z, Li H, Guo S. Salvianolic acid A inhibits tumor-associated angiogenesis by blocking GRP78 secretion. Naunyn Schmiedebergs Arch Pharmacol 2018; 392:467-480. [DOI: 10.1007/s00210-018-1585-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/12/2018] [Indexed: 12/31/2022]
|
43
|
Dougherty JA, Kumar N, Noor M, Angelos MG, Khan M, Chen CA, Khan M. Extracellular Vesicles Released by Human Induced-Pluripotent Stem Cell-Derived Cardiomyocytes Promote Angiogenesis. Front Physiol 2018; 9:1794. [PMID: 30618806 PMCID: PMC6302004 DOI: 10.3389/fphys.2018.01794] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 11/29/2018] [Indexed: 01/01/2023] Open
Abstract
Although cell survival post-transplantation is very low, emerging evidence using stem cell therapy for myocardial repair points toward a primary role of paracrine signaling mechanisms as the basis for improved cardiac function, decreased fibrosis, and increased angiogenesis. Recent studies have demonstrated that extracellular vesicles (EVs) such as exosomes secreted by stem cells stimulate angiogenesis, provide cytoprotection, and modulate apoptosis. However, the angiogenic potential of EVs secreted from human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM), a terminally differentiated cell type, has not been elucidated yet. Therefore, the main objective of this study is to isolate, characterize, and evaluate the in vitro angiogenic potential of EVs collected from hiPSC-CM conditioned media. The hiPSC-CM were cultured for 2 weeks and EVs were isolated from cell culture medium. Isolated EVs were characterized by transmission electron microscopy (TEM), nanoparticle tracking analysis, and immunoblotting. Furthermore, the angiogenic potential of these EVs was evaluated by tube formation, wound-healing, and cell-proliferation assays in bovine aortic endothelial cells (BAEC). In addition, gene expression levels of growth factors was evaluated in hiPSC-derived endothelial cells (hiPSC-EC) treated with hiPSC-CM-derived EV (CM-EVs) to assess their role in promoting angiogenesis. TEM imaging of CM-EVs showed a presence of a double-membrane bound structure, which is a characteristic of EV. Nanoparticle tracking analysis further confirmed the size and shape of the secreted particles to be consistent with EVs. Furthermore, EV-specific markers (CD63 and HSP70) were enriched in these particles as illustrated by immunoblotting. Most importantly, BAEC treated with 100 μg/ml of CM-EVs showed significant increases in tube formation, wound closure, and cell proliferation as compared to control (no-EVs). Finally, treatment of hiPSC-EC with CM-EVs induced increased expression of pro-angiogenic growth factors by the endothelial cells. Overall, our results demonstrated that EVs isolated from hiPSC-CM enhance angiogenesis in endothelial cells. This acellular/cell-free approach constitutes a potential translational therapeutic to induce angiogenesis in patients with myocardial infarction.
Collapse
Affiliation(s)
- Julie A Dougherty
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Naresh Kumar
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Mohammad Noor
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Mark G Angelos
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Mohsin Khan
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Chun-An Chen
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Mahmood Khan
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
44
|
Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, Antoniou A, Arab T, Archer F, Atkin-Smith GK, Ayre DC, Bach JM, Bachurski D, Baharvand H, Balaj L, Baldacchino S, Bauer NN, Baxter AA, Bebawy M, Beckham C, Bedina Zavec A, Benmoussa A, Berardi AC, Bergese P, Bielska E, Blenkiron C, Bobis-Wozowicz S, Boilard E, Boireau W, Bongiovanni A, Borràs FE, Bosch S, Boulanger CM, Breakefield X, Breglio AM, Brennan MÁ, Brigstock DR, Brisson A, Broekman MLD, Bromberg JF, Bryl-Górecka P, Buch S, Buck AH, Burger D, Busatto S, Buschmann D, Bussolati B, Buzás EI, Byrd JB, Camussi G, Carter DRF, Caruso S, Chamley LW, Chang YT, Chen C, Chen S, Cheng L, Chin AR, Clayton A, Clerici SP, Cocks A, Cocucci E, Coffey RJ, Cordeiro-da-Silva A, Couch Y, Coumans FAW, Coyle B, Crescitelli R, Criado MF, D’Souza-Schorey C, Das S, Datta Chaudhuri A, de Candia P, De Santana EF, De Wever O, del Portillo HA, Demaret T, Deville S, Devitt A, Dhondt B, Di Vizio D, Dieterich LC, Dolo V, Dominguez Rubio AP, Dominici M, Dourado MR, Driedonks TAP, Duarte FV, Duncan HM, Eichenberger RM, Ekström K, EL Andaloussi S, Elie-Caille C, Erdbrügger U, Falcón-Pérez JM, Fatima F, Fish JE, Flores-Bellver M, Försönits A, Frelet-Barrand A, Fricke F, Fuhrmann G, Gabrielsson S, Gámez-Valero A, Gardiner C, Gärtner K, Gaudin R, Gho YS, Giebel B, Gilbert C, Gimona M, Giusti I, Goberdhan DCI, Görgens A, Gorski SM, Greening DW, Gross JC, Gualerzi A, Gupta GN, Gustafson D, Handberg A, Haraszti RA, Harrison P, Hegyesi H, Hendrix A, Hill AF, Hochberg FH, Hoffmann KF, Holder B, Holthofer H, Hosseinkhani B, Hu G, Huang Y, Huber V, Hunt S, Ibrahim AGE, Ikezu T, Inal JM, Isin M, Ivanova A, Jackson HK, Jacobsen S, Jay SM, Jayachandran M, Jenster G, Jiang L, Johnson SM, Jones JC, Jong A, Jovanovic-Talisman T, Jung S, Kalluri R, Kano SI, Kaur S, Kawamura Y, Keller ET, Khamari D, Khomyakova E, Khvorova A, Kierulf P, Kim KP, Kislinger T, Klingeborn M, Klinke DJ, Kornek M, Kosanović MM, Kovács ÁF, Krämer-Albers EM, Krasemann S, Krause M, Kurochkin IV, Kusuma GD, Kuypers S, Laitinen S, Langevin SM, Languino LR, Lannigan J, Lässer C, Laurent LC, Lavieu G, Lázaro-Ibáñez E, Le Lay S, Lee MS, Lee YXF, Lemos DS, Lenassi M, Leszczynska A, Li ITS, Liao K, Libregts SF, Ligeti E, Lim R, Lim SK, Linē A, Linnemannstöns K, Llorente A, Lombard CA, Lorenowicz MJ, Lörincz ÁM, Lötvall J, Lovett J, Lowry MC, Loyer X, Lu Q, Lukomska B, Lunavat TR, Maas SLN, Malhi H, Marcilla A, Mariani J, Mariscal J, Martens-Uzunova ES, Martin-Jaular L, Martinez MC, Martins VR, Mathieu M, Mathivanan S, Maugeri M, McGinnis LK, McVey MJ, Meckes DG, Meehan KL, Mertens I, Minciacchi VR, Möller A, Møller Jørgensen M, Morales-Kastresana A, Morhayim J, Mullier F, Muraca M, Musante L, Mussack V, Muth DC, Myburgh KH, Najrana T, Nawaz M, Nazarenko I, Nejsum P, Neri C, Neri T, Nieuwland R, Nimrichter L, Nolan JP, Nolte-’t Hoen ENM, Noren Hooten N, O’Driscoll L, O’Grady T, O’Loghlen A, Ochiya T, Olivier M, Ortiz A, Ortiz LA, Osteikoetxea X, Østergaard O, Ostrowski M, Park J, Pegtel DM, Peinado H, Perut F, Pfaffl MW, Phinney DG, Pieters BCH, Pink RC, Pisetsky DS, Pogge von Strandmann E, Polakovicova I, Poon IKH, Powell BH, Prada I, Pulliam L, Quesenberry P, Radeghieri A, Raffai RL, Raimondo S, Rak J, Ramirez MI, Raposo G, Rayyan MS, Regev-Rudzki N, Ricklefs FL, Robbins PD, Roberts DD, Rodrigues SC, Rohde E, Rome S, Rouschop KMA, Rughetti A, Russell AE, Saá P, Sahoo S, Salas-Huenuleo E, Sánchez C, Saugstad JA, Saul MJ, Schiffelers RM, Schneider R, Schøyen TH, Scott A, Shahaj E, Sharma S, Shatnyeva O, Shekari F, Shelke GV, Shetty AK, Shiba K, Siljander PRM, Silva AM, Skowronek A, Snyder OL, Soares RP, Sódar BW, Soekmadji C, Sotillo J, Stahl PD, Stoorvogel W, Stott SL, Strasser EF, Swift S, Tahara H, Tewari M, Timms K, Tiwari S, Tixeira R, Tkach M, Toh WS, Tomasini R, Torrecilhas AC, Tosar JP, Toxavidis V, Urbanelli L, Vader P, van Balkom BWM, van der Grein SG, Van Deun J, van Herwijnen MJC, Van Keuren-Jensen K, van Niel G, van Royen ME, van Wijnen AJ, Vasconcelos MH, Vechetti IJ, Veit TD, Vella LJ, Velot É, Verweij FJ, Vestad B, Viñas JL, Visnovitz T, Vukman KV, Wahlgren J, Watson DC, Wauben MHM, Weaver A, Webber JP, Weber V, Wehman AM, Weiss DJ, Welsh JA, Wendt S, Wheelock AM, Wiener Z, Witte L, Wolfram J, Xagorari A, Xander P, Xu J, Yan X, Yáñez-Mó M, Yin H, Yuana Y, Zappulli V, Zarubova J, Žėkas V, Zhang JY, Zhao Z, Zheng L, Zheutlin AR, Zickler AM, Zimmermann P, Zivkovic AM, Zocco D, Zuba-Surma EK. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 2018; 7:1535750. [PMID: 30637094 PMCID: PMC6322352 DOI: 10.1080/20013078.2018.1535750] [Citation(s) in RCA: 6864] [Impact Index Per Article: 1144.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 09/25/2018] [Indexed: 11/04/2022] Open
Abstract
The last decade has seen a sharp increase in the number of scientific publications describing physiological and pathological functions of extracellular vesicles (EVs), a collective term covering various subtypes of cell-released, membranous structures, called exosomes, microvesicles, microparticles, ectosomes, oncosomes, apoptotic bodies, and many other names. However, specific issues arise when working with these entities, whose size and amount often make them difficult to obtain as relatively pure preparations, and to characterize properly. The International Society for Extracellular Vesicles (ISEV) proposed Minimal Information for Studies of Extracellular Vesicles ("MISEV") guidelines for the field in 2014. We now update these "MISEV2014" guidelines based on evolution of the collective knowledge in the last four years. An important point to consider is that ascribing a specific function to EVs in general, or to subtypes of EVs, requires reporting of specific information beyond mere description of function in a crude, potentially contaminated, and heterogeneous preparation. For example, claims that exosomes are endowed with exquisite and specific activities remain difficult to support experimentally, given our still limited knowledge of their specific molecular machineries of biogenesis and release, as compared with other biophysically similar EVs. The MISEV2018 guidelines include tables and outlines of suggested protocols and steps to follow to document specific EV-associated functional activities. Finally, a checklist is provided with summaries of key points.
Collapse
Affiliation(s)
- Clotilde Théry
- Institut Curie, INSERM U932, PSL Research University, Paris, France
| | - Kenneth W Witwer
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
- The Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, USA
| | - Elena Aikawa
- Brigham and Women’s Hospital, Center for Interdisciplinary Cardiovascular Sciences, Boston, MA, USA
- Harvard Medical School, Cardiovascular Medicine, Boston, MA, USA
| | - Maria Jose Alcaraz
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM), University of Valencia, Polytechnic University of Valencia, Valencia, Spain
| | | | | | - Anna Antoniou
- German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University Hospital Bonn (UKB), Bonn, Germany
| | - Tanina Arab
- Université de Lille, INSERM, U-1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse - PRISM, Lille, France
| | - Fabienne Archer
- University of Lyon, INRA, EPHE, UMR754 Viral Infections and Comparative Pathology, Lyon, France
| | - Georgia K Atkin-Smith
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - D Craig Ayre
- Atlantic Cancer Research Institute, Moncton, Canada
- Mount Allison University, Department of Chemistry and Biochemistry, Sackville, Canada
| | - Jean-Marie Bach
- Université Bretagne Loire, Oniris, INRA, IECM, Nantes, France
| | - Daniel Bachurski
- University of Cologne, Department of Internal Medicine I, Cologne, Germany
| | - Hossein Baharvand
- Royan Institute for Stem Cell Biology and Technology, ACECR, Cell Science Research Center, Department of Stem Cells and Developmental Biology, Tehran, Iran
- University of Science and Culture, ACECR, Department of Developmental Biology, Tehran, Iran
| | - Leonora Balaj
- Massachusetts General Hospital, Department of Neurosurgery, Boston, MA, USA
| | | | - Natalie N Bauer
- University of South Alabama, Department of Pharmacology, Center for Lung Biology, Mobile, AL, USA
| | - Amy A Baxter
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Mary Bebawy
- University of Technology Sydney, Discipline of Pharmacy, Graduate School of Health, Sydney, Australia
| | | | - Apolonija Bedina Zavec
- National Institute of Chemistry, Department of Molecular Biology and Nanobiotechnology, Ljubljana, Slovenia
| | - Abderrahim Benmoussa
- Université Laval, Centre de Recherche du CHU de Québec, Department of Infectious Diseases and Immunity, Quebec City, Canada
| | | | - Paolo Bergese
- CSGI - Research Center for Colloids and Nanoscience, Florence, Italy
- INSTM - National Interuniversity Consortium of Materials Science and Technology, Florence, Italy
- University of Brescia, Department of Molecular and Translational Medicine, Brescia, Italy
| | - Ewa Bielska
- University of Birmingham, Institute of Microbiology and Infection, Birmingham, UK
| | | | - Sylwia Bobis-Wozowicz
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Kraków, Poland
| | - Eric Boilard
- Université Laval, Centre de Recherche du CHU de Québec, Department of Infectious Diseases and Immunity, Quebec City, Canada
| | - Wilfrid Boireau
- FEMTO-ST Institute, UBFC, CNRS, ENSMM, UTBM, Besançon, France
| | - Antonella Bongiovanni
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council (CNR) of Italy, Palermo, Italy
| | - Francesc E Borràs
- Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, REMAR-IVECAT Group, Badalona, Spain
- Germans Trias i Pujol University Hospital, Nephrology Service, Badalona, Spain
- Universitat Autònoma de Barcelona, Department of Cell Biology, Physiology & Immunology, Barcelona, Spain
| | - Steffi Bosch
- Université Bretagne Loire, Oniris, INRA, IECM, Nantes, France
| | - Chantal M Boulanger
- INSERM UMR-S 970, Paris Cardiovascular Research Center, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Xandra Breakefield
- Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, Department of Neurology and Radiology, Boston, MA, USA
| | - Andrew M Breglio
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- National Institutes of Health, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, USA
| | - Meadhbh Á Brennan
- Harvard University, School of Engineering and Applied Sciences, Cambridge, MA, USA
- Massachusetts General Hospital, Harvard Medical School, Department of Neurology, Boston, MA, USA
- Université de Nantes, INSERM UMR 1238, Bone Sarcoma and Remodeling of Calcified Tissues, PhyOS, Nantes, France
| | - David R Brigstock
- Nationwide Children’s Hospital, Columbus, OH, USA
- The Ohio State University, Columbus, OH, USA
| | - Alain Brisson
- UMR-CBMN, CNRS-Université de Bordeaux, Bordeaux, France
| | - Marike LD Broekman
- Haaglanden Medical Center, Department of Neurosurgery, The Hague, The Netherlands
- Leiden University Medical Center, Department of Neurosurgery, Leiden, The Netherlands
- Massachusetts General Hospital, Department of Neurology, Boston, MA, USA
| | - Jacqueline F Bromberg
- Memorial Sloan Kettering Cancer Center, Department of Medicine, New York City, NY, USA
- Weill Cornell Medicine, Department of Medicine, New York City, NY, USA
| | | | - Shilpa Buch
- University of Nebraska Medical Center, Department of Pharmacology and Experimental Neuroscience, Omaha, NE, USA
| | - Amy H Buck
- University of Edinburgh, Institute of Immunology & Infection Research, Edinburgh, UK
| | - Dylan Burger
- Kidney Research Centre, Ottawa, Canada
- Ottawa Hospital Research Institute, Ottawa, Canada
- University of Ottawa, Ottawa, Canada
| | - Sara Busatto
- Mayo Clinic, Department of Transplantation, Jacksonville, FL, USA
- University of Brescia, Department of Molecular and Translational Medicine, Brescia, Italy
| | - Dominik Buschmann
- Technical University of Munich, TUM School of Life Sciences Weihenstephan, Division of Animal Physiology and Immunology, Freising, Germany
| | - Benedetta Bussolati
- University of Torino, Department of Molecular Biotechnology and Health Sciences, Torino, Italy
| | - Edit I Buzás
- MTA-SE Immuno-Proteogenomics Research Groups, Budapest, Hungary
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - James Bryan Byrd
- University of Michigan, Department of Medicine, Ann Arbor, MI, USA
| | - Giovanni Camussi
- University of Torino, Department of Medical Sciences, Torino, Italy
| | - David RF Carter
- Oxford Brookes University, Department of Biological and Medical Sciences, Oxford, UK
| | - Sarah Caruso
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Lawrence W Chamley
- University of Auckland, Department of Obstetrics and Gynaecology, Auckland, New Zealand
| | - Yu-Ting Chang
- National Taiwan University Hospital, Department of Internal Medicine, Taipei, Taiwan
| | - Chihchen Chen
- National Tsing Hua University, Department of Power Mechanical Engineering, Hsinchu, Taiwan
- National Tsing Hua University, Institute of Nanoengineering and Microsystems, Hsinchu, Taiwan
| | - Shuai Chen
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Reproductive Biology, Dummerstorf, Germany
| | - Lesley Cheng
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | | | - Aled Clayton
- Cardiff University, School of Medicine, Cardiff, UK
| | | | - Alex Cocks
- Cardiff University, School of Medicine, Cardiff, UK
| | - Emanuele Cocucci
- The Ohio State University, College of Pharmacy, Division of Pharmaceutics and Pharmaceutical Chemistry, Columbus, OH, USA
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH, USA
| | - Robert J Coffey
- Vanderbilt University Medical Center, Epithelial Biology Center, Department of Medicine, Nashville, TN, USA
| | | | - Yvonne Couch
- University of Oxford, Radcliffe Department of Medicine, Acute Stroke Programme - Investigative Medicine, Oxford, UK
| | - Frank AW Coumans
- Academic Medical Centre of the University of Amsterdam, Department of Clinical Chemistry and Vesicle Observation Centre, Amsterdam, The Netherlands
| | - Beth Coyle
- The University of Nottingham, School of Medicine, Children’s Brain Tumour Research Centre, Nottingham, UK
| | - Rossella Crescitelli
- University of Gothenburg, Institute of Medicine at Sahlgrenska Academy, Krefting Research Centre, Gothenburg, Sweden
| | | | | | - Saumya Das
- Massachusetts General Hospital, Boston, MA, USA
| | - Amrita Datta Chaudhuri
- The Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, USA
| | | | - Eliezer F De Santana
- The Sociedade Beneficente Israelita Brasileira Albert Einstein, São Paulo, Brazil
| | - Olivier De Wever
- Cancer Research Institute Ghent, Ghent, Belgium
- Ghent University, Department of Radiation Oncology and Experimental Cancer Research, Laboratory of Experimental Cancer Research, Ghent, Belgium
| | - Hernando A del Portillo
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Institut d’Investigació Germans Trias i Pujol (IGTP), PVREX group, Badalona, Spain
- ISGlobal, Hospital Clínic - Universitat de Barcelona, PVREX Group, Barcelona, Spain
| | - Tanguy Demaret
- Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Brussels, Belgium
| | - Sarah Deville
- Universiteit Hasselt, Diepenbeek, Belgium
- Vlaamse Instelling voor Technologisch Onderzoek (VITO), Mol, Belgium
| | - Andrew Devitt
- Aston University, School of Life & Health Sciences, Birmingham, UK
| | - Bert Dhondt
- Cancer Research Institute Ghent, Ghent, Belgium
- Ghent University Hospital, Department of Urology, Ghent, Belgium
- Ghent University, Department of Radiation Oncology and Experimental Cancer Research, Laboratory of Experimental Cancer Research, Ghent, Belgium
| | | | | | - Vincenza Dolo
- University of L’Aquila, Department of Life, Health and Environmental Sciences, L’Aquila, Italy
| | - Ana Paula Dominguez Rubio
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Buenos Aires, Argentina
| | - Massimo Dominici
- TPM of Mirandola, Mirandola, Italy
- University of Modena and Reggio Emilia, Division of Oncology, Modena, Italy
| | - Mauricio R Dourado
- University of Campinas, Piracicaba Dental School, Department of Oral Diagnosis, Piracicaba, Brazil
- University of Oulu, Faculty of Medicine, Cancer and Translational Medicine Research Unit, Oulu, Finland
| | - Tom AP Driedonks
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | | | - Heather M Duncan
- McGill University, Division of Experimental Medicine, Montreal, Canada
- McGill University, The Research Institute of the McGill University Health Centre, Child Health and Human Development Program, Montreal, Canada
| | - Ramon M Eichenberger
- James Cook University, Australian Institute of Tropical Health and Medicine, Centre for Biodiscovery and Molecular Development of Therapeutics, Cairns, Australia
| | - Karin Ekström
- University of Gothenburg, Institute of Clinical Sciences at Sahlgrenska Academy, Department of Biomaterials, Gothenburg, Sweden
| | - Samir EL Andaloussi
- Evox Therapeutics Limited, Oxford, UK
- Karolinska Institute, Stockholm, Sweden
| | | | - Uta Erdbrügger
- University of Virginia Health System, Department of Medicine, Division of Nephrology, Charlottesville, VA, USA
| | - Juan M Falcón-Pérez
- CIC bioGUNE, CIBERehd, Exosomes Laboratory & Metabolomics Platform, Derio, Spain
- IKERBASQUE Research Science Foundation, Bilbao, Spain
| | - Farah Fatima
- University of São Paulo, Ribeirão Preto Medical School, Department of Pathology and Forensic Medicine, Ribeirão Preto, Brazil
| | - Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- University of Toronto, Department of Laboratory Medicine and Pathobiology, Toronto, Canada
| | - Miguel Flores-Bellver
- University of Colorado, School of Medicine, Department of Ophthalmology, Cell Sight-Ocular Stem Cell and Regeneration Program, Aurora, CO, USA
| | - András Försönits
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | | | - Fabia Fricke
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Applied Tumor Biology, Heidelberg, Germany
- University Hospital Heidelberg, Institute of Pathology, Applied Tumor Biology, Heidelberg, Germany
| | - Gregor Fuhrmann
- Helmholtz-Centre for Infection Research, Braunschweig, Germany
- Helmholtz-Institute for Pharmaceutical Research Saarland, Saarbrücken, Germany
- Saarland University, Saarbrücken, Germany
| | - Susanne Gabrielsson
- Karolinska Institute, Department of Medicine Solna, Division for Immunology and Allergy, Stockholm, Sweden
| | - Ana Gámez-Valero
- Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, REMAR-IVECAT Group, Badalona, Spain
- Universitat Autònoma de Barcelona, Hospital Universitari and Health Sciences Research Institute Germans Trias i Pujol, Department of Pathology, Barcelona, Spain
| | | | - Kathrin Gärtner
- Helmholtz Center Munich German Research Center for Environmental Health, Research Unit Gene Vectors, Munich, Germany
| | - Raphael Gaudin
- INSERM U1110, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Yong Song Gho
- POSTECH (Pohang University of Science and Technology), Department of Life Sciences, Pohang, South Korea
| | - Bernd Giebel
- University Hospital Essen, University Duisburg-Essen, Institute for Transfusion Medicine, Essen, Germany
| | - Caroline Gilbert
- Université Laval, Centre de Recherche du CHU de Québec, Department of Infectious Diseases and Immunity, Quebec City, Canada
| | - Mario Gimona
- Paracelsus Medical University, GMP Unit, Salzburg, Austria
| | - Ilaria Giusti
- University of L’Aquila, Department of Life, Health and Environmental Sciences, L’Aquila, Italy
| | - Deborah CI Goberdhan
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, UK
| | - André Görgens
- Evox Therapeutics Limited, Oxford, UK
- Karolinska Institute, Clinical Research Center, Department of Laboratory Medicine, Stockholm, Sweden
- University Hospital Essen, University Duisburg-Essen, Institute for Transfusion Medicine, Essen, Germany
| | - Sharon M Gorski
- BC Cancer, Canada’s Michael Smith Genome Sciences Centre, Vancouver, Canada
- Simon Fraser University, Department of Molecular Biology and Biochemistry, Burnaby, Canada
| | - David W Greening
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Julia Christina Gross
- University Medical Center Göttingen, Developmental Biochemistry, Göttingen, Germany
- University Medical Center Göttingen, Hematology and Oncology, Göttingen, Germany
| | - Alice Gualerzi
- IRCCS Fondazione Don Carlo Gnocchi, Laboratory of Nanomedicine and Clinical Biophotonics (LABION), Milan, Italy
| | - Gopal N Gupta
- Loyola University Chicago, Department of Urology, Maywood, IL, USA
| | - Dakota Gustafson
- University of Toronto, Department of Laboratory Medicine and Pathobiology, Toronto, Canada
| | - Aase Handberg
- Aalborg University Hospital, Department of Clinical Biochemistry, Aalborg, Denmark
- Aalborg University, Clinical Institute, Aalborg, Denmark
| | - Reka A Haraszti
- University of Massachusetts Medical School, RNA Therapeutics Institute, Worcester, MA, USA
| | | | - Hargita Hegyesi
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - An Hendrix
- Cancer Research Institute Ghent, Ghent, Belgium
- Ghent University, Department of Radiation Oncology and Experimental Cancer Research, Laboratory of Experimental Cancer Research, Ghent, Belgium
| | - Andrew F Hill
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Fred H Hochberg
- Scintillon Institute, La Jolla, CA, USA
- University of California, San Diego, Department of Neurosurgery, La Jolla, CA, USA
| | - Karl F Hoffmann
- Aberystwyth University, Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth, United Kingdom
| | - Beth Holder
- Imperial College London, London, UK
- MRC The Gambia, Fajara, The Gambia
| | | | - Baharak Hosseinkhani
- Hasselt University, Biomedical Research Institute (BIOMED), Department of Medicine and Life Sciences, Hasselt, Belgium
| | - Guoku Hu
- University of Nebraska Medical Center, Department of Pharmacology and Experimental Neuroscience, Omaha, NE, USA
| | - Yiyao Huang
- Nanfang Hospital, Southern Medical University, Department of Clinical Laboratory Medicine, Guangzhou, China
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | - Veronica Huber
- Fondazione IRCCS Istituto Nazionale dei Tumori, Unit of Immunotherapy of Human Tumors, Milan, Italy
| | | | | | - Tsuneya Ikezu
- Boston University School of Medicine, Boston, MA, USA
| | - Jameel M Inal
- University of Hertfordshire, School of Life and Medical Sciences, Biosciences Research Group, Hatfield, UK
| | - Mustafa Isin
- Istanbul University Oncology Institute, Basic Oncology Department, Istanbul, Turkey
| | - Alena Ivanova
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg, Germany
| | - Hannah K Jackson
- The University of Nottingham, School of Medicine, Children’s Brain Tumour Research Centre, Nottingham, UK
| | - Soren Jacobsen
- Copenhagen Lupus and Vasculitis Clinic, Section 4242 - Rigshospitalet, Copenhagen, Denmark
- University of Copenhagen, Institute of Clinical Medicine, Copenhagen, Denmark
| | - Steven M Jay
- University of Maryland, Fischell Department of Bioengineering, College Park, MD, USA
| | - Muthuvel Jayachandran
- Mayo Clinic, College of Medicine, Department of Physiology and Biomedical Engineering, Rochester, MN, USA
| | | | - Lanzhou Jiang
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Suzanne M Johnson
- University of Manchester, Division of Cancer Sciences, Manchester Cancer Research Centre, Manchester, UK
| | - Jennifer C Jones
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, MD, USA
| | - Ambrose Jong
- Children’s Hospital of Los Angeles, Los Angeles, CA, USA
- University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Tijana Jovanovic-Talisman
- City of Hope Comprehensive Cancer Center, Beckman Research Institute, Department of Molecular Medicine, Duarte, CA, USA
| | - Stephanie Jung
- German Research Center for Environmental Health, Institute for Virology, Munich, Germany
| | - Raghu Kalluri
- University of Texas MD Anderson Cancer Center, Department of Cancer Biology, Metastasis Research Center, Houston, TX, USA
| | - Shin-ichi Kano
- The Johns Hopkins University School of Medicine, Department of Psychiatry and Behavioral Sciences, Baltimore, MD, USA
| | - Sukhbir Kaur
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Laboratory of Pathology, Bethesda, MD, USA
| | - Yumi Kawamura
- National Cancer Center Research Institute, Tokyo, Japan
- University of Tsukuba, Tsukuba, Japan
| | - Evan T Keller
- University of Michigan, Biointerfaces Institute, Ann Arbor, MI, USA
- University of Michigan, Department of Urology, Ann Arbor, MI, USA
| | - Delaram Khamari
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - Elena Khomyakova
- École normale supérieure, Paris, France
- Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow, Russia
| | - Anastasia Khvorova
- University of Massachusetts Medical School, RNA Therapeutics Institute, Worcester, MA, USA
| | - Peter Kierulf
- Oslo University Hospital, Department of Medical Biochemistry, Blood Cell Research Group, Oslo, Norway
| | - Kwang Pyo Kim
- Kyung Hee University, Department of Applied Chemistry, Yongin, Korea
| | - Thomas Kislinger
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- University of Toronto, Department of Medical Biophysics, Toronto, Canada
| | | | - David J Klinke
- West Virginia University, Department of Chemical and Biomedical Engineering and WVU Cancer Institute, Morgantown, WV, USA
- West Virginia University, Department of Microbiology Immunology and Cell Biology, Morgantown, WV, USA
| | - Miroslaw Kornek
- German Armed Forces Central Hospital, Department of General, Visceral and Thoracic Surgery, Koblenz, Germany
- Saarland University Medical Center, Department of Medicine II, Homburg, Germany
| | - Maja M Kosanović
- University of Belgrade, Institute for the Application of Nuclear Energy, INEP, Belgrade, Serbia
| | - Árpád Ferenc Kovács
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | | | - Susanne Krasemann
- University Medical Center Hamburg-Eppendorf, Institute of Neuropathology, Hamburg, Germany
| | - Mirja Krause
- Hudson Institute of Medical Research, Melbourne, Australia
| | | | - Gina D Kusuma
- Hudson Institute of Medical Research, Melbourne, Australia
- Monash University, Melbourne, Australia
| | - Sören Kuypers
- Hasselt University, Biomedical Research Institute (BIOMED), Hasselt, Belgium
| | - Saara Laitinen
- Finnish Red Cross Blood Service, Research and Development, Helsinki, Finland
| | - Scott M Langevin
- Cincinnati Cancer Center, Cincinnati, OH, USA
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lucia R Languino
- Thomas Jefferson University, Sidney Kimmel Medical School, Department of Cancer Biology, Philadelphia, PA, USA
| | - Joanne Lannigan
- University of Virginia, Flow Cytometry Core, School of Medicine, Charlottesville, VA, USA
| | - Cecilia Lässer
- University of Gothenburg, Institute of Medicine at Sahlgrenska Academy, Krefting Research Centre, Gothenburg, Sweden
| | - Louise C Laurent
- University of California, San Diego, Department of Obstetrics, Gynecology, and Reproductive Sciences, La Jolla, CA, USA
| | - Gregory Lavieu
- Institut Curie, INSERM U932, PSL Research University, Paris, France
| | | | - Soazig Le Lay
- INSERM U1063, Université d’Angers, CHU d’Angers, Angers, France
| | - Myung-Shin Lee
- Eulji University, School of Medicine, Daejeon, South Korea
| | | | - Debora S Lemos
- Federal University of Paraná, Department of Genetics, Human Molecular Genetics Laboratory, Curitiba, Brazil
| | - Metka Lenassi
- University of Ljubljana, Faculty of Medicine, Institute of Biochemistry, Ljubljana, Slovenia
| | | | - Isaac TS Li
- University of British Columbia Okanagan, Kelowna, Canada
| | - Ke Liao
- University of Nebraska Medical Center, Department of Pharmacology and Experimental Neuroscience, Omaha, NE, USA
| | - Sten F Libregts
- University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Department of Medicine, Cambridge NIHR BRC Cell Phenotyping Hub, Cambridge, UK
| | - Erzsebet Ligeti
- Semmelweis University, Department of Physiology, Budapest, Hungary
| | - Rebecca Lim
- Hudson Institute of Medical Research, Melbourne, Australia
- Monash University, Melbourne, Australia
| | - Sai Kiang Lim
- Institute of Medical Biology (IMB), Agency for Science and Technology (A*STAR), Singapore
| | - Aija Linē
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Karen Linnemannstöns
- University Medical Center Göttingen, Developmental Biochemistry, Göttingen, Germany
- University Medical Center Göttingen, Hematology and Oncology, Göttingen, Germany
| | - Alicia Llorente
- Oslo University Hospital-The Norwegian Radium Hospital, Institute for Cancer Research, Department of Molecular Cell Biology, Oslo, Norway
| | - Catherine A Lombard
- Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Brussels, Belgium
| | - Magdalena J Lorenowicz
- Utrecht University, University Medical Center Utrecht, Center for Molecular Medicine & Regenerative Medicine Center, Utrecht, The Netherlands
| | - Ákos M Lörincz
- Semmelweis University, Department of Physiology, Budapest, Hungary
| | - Jan Lötvall
- University of Gothenburg, Institute of Medicine at Sahlgrenska Academy, Krefting Research Centre, Gothenburg, Sweden
| | - Jason Lovett
- Stellenbosch University, Department of Physiological Sciences, Stellenbosch, South Africa
| | - Michelle C Lowry
- Trinity College Dublin, School of Pharmacy and Pharmaceutical Sciences, Panoz Institute & Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Xavier Loyer
- INSERM UMR-S 970, Paris Cardiovascular Research Center, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Quan Lu
- Harvard University, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Barbara Lukomska
- Mossakowski Medical Research Centre, NeuroRepair Department, Warsaw, Poland
| | - Taral R Lunavat
- K.G. Jebsen Brain Tumor Research Centre, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Sybren LN Maas
- Utrecht University, University Medical Center Utrecht, Department of Neurosurgery, Brain Center Rudolf Magnus, Institute of Neurosciences, Utrecht, The Netherlands
- Utrecht University, University Medical Center Utrecht, Department of Pathology, Utrecht, The Netherlands
| | | | - Antonio Marcilla
- Universitat de València, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Àrea de Parasitologia, Valencia, Spain
- Universitat de València, Health Research Institute La Fe, Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Valencia, Spain
| | - Jacopo Mariani
- Università degli Studi di Milano, Department of Clinical Sciences and Community Health, EPIGET LAB, Milan, Italy
| | | | | | | | | | | | - Mathilde Mathieu
- Institut Curie, INSERM U932, PSL Research University, Paris, France
| | - Suresh Mathivanan
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Marco Maugeri
- University of Gothenburg, Sahlgrenska Academy, Department of Rheumatology and Inflammation Research, Gothenburg, Sweden
| | | | - Mark J McVey
- SickKids Hospital, Department of Anesthesia and Pain Medicine, Toronto, Canada
- University of Toronto, Department of Anesthesia, Toronto, Canada
| | - David G Meckes
- Florida State University College of Medicine, Department of Biomedical Sciences, Tallahassee, FL, USA
| | - Katie L Meehan
- The School of Biomedical Sciences, University of Western Australia, Perth, Australia
| | - Inge Mertens
- University of Antwerp, Centre for Proteomics, Antwerp, Belgium
- Vlaamse Instelling voor Technologisch Onderzoek (VITO), Mol, Belgium
| | - Valentina R Minciacchi
- Georg-Speyer-Haus Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Andreas Möller
- QIMR Berghofer Medical Research Institute, Herston, Australia
| | - Malene Møller Jørgensen
- Aalborg University Hospital, Department of Clinical Immunology, Aalborg, Denmark
- EVSEARCH.DK, Denmark
| | - Aizea Morales-Kastresana
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, MD, USA
| | | | - François Mullier
- Namur Thrombosis and Hemostasis Center (NTHC), NARILIS, Namur, Belgium
- Université Catholique de Louvain, CHU UCL Namur, Hematology-Hemostasis Laboratory, Yvoir, Belgium
| | - Maurizio Muraca
- University of Padova, Department of Women’s and Children’s Health, Padova, Italy
| | - Luca Musante
- University of Virginia Health System, Department of Medicine, Division of Nephrology, Charlottesville, VA, USA
| | - Veronika Mussack
- Technical University of Munich, TUM School of Life Sciences Weihenstephan, Division of Animal Physiology and Immunology, Freising, Germany
| | - Dillon C Muth
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | - Kathryn H Myburgh
- Stellenbosch University, Department of Physiological Sciences, Stellenbosch, South Africa
| | - Tanbir Najrana
- Brown University, Women and Infants Hospital, Providence, RI, USA
| | - Muhammad Nawaz
- University of Gothenburg, Sahlgrenska Academy, Department of Rheumatology and Inflammation Research, Gothenburg, Sweden
| | - Irina Nazarenko
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Institute for Infection Prevention and Hospital Epidemiology, Freiburg, Germany
| | - Peter Nejsum
- Aarhus University, Department of Clinical Medicine, Aarhus, Denmark
| | - Christian Neri
- Sorbonne Université, Centre National de la Recherche Scientifique, Research Unit Biology of Adaptation and Aging (B2A), Team Compensation in Neurodegenerative and Aging (Brain-C), Paris, France
| | - Tommaso Neri
- University of Pisa, Centro Dipartimentale di Biologia Cellulare Cardio-Respiratoria, Pisa, Italy
| | - Rienk Nieuwland
- Academic Medical Centre of the University of Amsterdam, Department of Clinical Chemistry and Vesicle Observation Centre, Amsterdam, The Netherlands
| | - Leonardo Nimrichter
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia, Rio de Janeiro, Brazil
| | | | - Esther NM Nolte-’t Hoen
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | - Nicole Noren Hooten
- National Institutes of Health, National Institute on Aging, Baltimore, MD, USA
| | - Lorraine O’Driscoll
- Trinity College Dublin, School of Pharmacy and Pharmaceutical Sciences, Panoz Institute & Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Tina O’Grady
- University of Liège, GIGA-R(MBD), PSI Laboratory, Liège, Belgium
| | - Ana O’Loghlen
- Queen Mary University of London, Blizard Institute, Epigenetics & Cellular Senescence Group, London, UK
| | - Takahiro Ochiya
- National Cancer Center Research Institute, Division of Molecular and Cellular Medicine, Tokyo, Japan
| | - Martin Olivier
- McGill University, The Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Alberto Ortiz
- IIS-Fundacion Jimenez Diaz-UAM, Department of Nephrology and Hypertension, Madrid, Spain
- Spanish Kidney Research Network, REDINREN, Madrid, Spain
- Universidad Autónoma de Madrid, School of Medicine, Department of Medicine, Madrid, Spain
| | - Luis A Ortiz
- Graduate School of Public Health at the University of Pittsburgh, Division of Occupational and Environmental Medicine, Pittsburgh, PA, USA
| | | | - Ole Østergaard
- Statens Serum Institut, Department of Autoimmunology and Biomarkers, Copenhagen, Denmark
- University of Copenhagen, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, Copenhagen, Denmark
| | - Matias Ostrowski
- University of Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
| | - Jaesung Park
- POSTECH (Pohang University of Science and Technology), Department of Life Sciences, Pohang, South Korea
| | - D. Michiel Pegtel
- Amsterdam University Medical Centers, Department of Pathology, Amsterdam, The Netherlands
| | - Hector Peinado
- Spanish National Cancer Research Center (CNIO), Molecular Oncology Programme, Microenvironment and Metastasis Laboratory, Madrid, Spain
| | - Francesca Perut
- IRCCS - Istituto Ortopedico Rizzoli, Laboratory for Orthopaedic Pathophysiology and Regenerative Medicine, Bologna, Italy
| | - Michael W Pfaffl
- Technical University of Munich, TUM School of Life Sciences Weihenstephan, Division of Animal Physiology and Immunology, Freising, Germany
| | - Donald G Phinney
- The Scripps Research Institute-Scripps Florida, Department of Molecular Medicine, Jupiter, FL, USA
| | - Bartijn CH Pieters
- Radboud University Medical Center, Department of Rheumatology, Nijmegen, The Netherlands
| | - Ryan C Pink
- Oxford Brookes University, Department of Biological and Medical Sciences, Oxford, UK
| | - David S Pisetsky
- Duke University Medical Center, Departments of Medicine and Immunology, Durham, NC, USA
- Durham VAMC, Medical Research Service, Durham, NC, USA
| | | | - Iva Polakovicova
- Pontificia Universidad Católica de Chile, Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Pontificia Universidad Católica de Chile, Faculty of Medicine, Department of Hematology-Oncology, Santiago, Chile
| | - Ivan KH Poon
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Bonita H Powell
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | | | - Lynn Pulliam
- University of California, San Francisco, CA, USA
- Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Peter Quesenberry
- The Warren Alpert Medical School of Brown University, Department of Medicine, Providence, RI, USA
| | - Annalisa Radeghieri
- CSGI - Research Center for Colloids and Nanoscience, Florence, Italy
- University of Brescia, Department of Molecular and Translational Medicine, Brescia, Italy
| | - Robert L Raffai
- Department of Veterans Affairs, San Francisco, CA, USA
- University of California, San Francisco, CA, USA
| | - Stefania Raimondo
- University of Palermo, Department of Biopathology and Medical Biotechnologies, Palermo, Italy
| | - Janusz Rak
- McGill University, Montreal, Canada
- McGill University, The Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Marcel I Ramirez
- Instituto Oswaldo Cruz, Rio de Janeiro, Brazil
- Universidade Federal de Paraná, Paraná, Brazil
| | - Graça Raposo
- Institut Curie, CNRS UMR144, PSL Research University, Paris, France
| | - Morsi S Rayyan
- University of Michigan Medical School, Ann Arbor, MI, USA
| | - Neta Regev-Rudzki
- Weizmann Institute of Science, Department of Biomolecular Sciences, Rehovot, Israel
| | - Franz L Ricklefs
- University Medical Center Hamburg-Eppendorf, Department of Neurosurgery, Hamburg, Germany
| | - Paul D Robbins
- University of Minnesota Medical School, Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, Minneapolis, MN, USA
| | - David D Roberts
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Laboratory of Pathology, Bethesda, MD, USA
| | | | - Eva Rohde
- Paracelsus Medical University, Department of Transfusion Medicine, Salzburg, Austria
- Paracelsus Medical University, GMP Unit, Salzburg, Austria
- Spinal Cord Injury & Tissue Regeneration Center Salzburg (SCI-TReCS), Salzburg, Austria
| | - Sophie Rome
- University of Lyon, Lyon-Sud Faculty of Medicine, CarMeN Laboratory (UMR INSERM 1060-INRA 1397), Pierre-Bénite, France
| | - Kasper MA Rouschop
- Maastricht University, GROW, School for Oncology and Developmental Biology, Maastricht Radiation Oncology (MaastRO) Lab, Maastricht, The Netherlands
| | - Aurelia Rughetti
- Sapienza University of Rome, Department of Experimental Medicine, Rome, Italy
| | | | - Paula Saá
- American Red Cross, Scientific Affairs, Gaithersburg, MD, USA
| | - Susmita Sahoo
- Icahn School of Medicine at Mount Sinai, Department of Medicine, Cardiology, New York City, NY, USA
| | - Edison Salas-Huenuleo
- Advanced Center for Chronic Diseases, Santiago, Chile
- University of Chile, Faculty of Chemical and Pharmaceutical Science, Laboratory of Nanobiotechnology and Nanotoxicology, Santiago, Chile
| | - Catherine Sánchez
- Clínica las Condes, Extracellular Vesicles in Personalized Medicine Group, Santiago, Chile
| | - Julie A Saugstad
- Oregon Health & Science University, Department of Anesthesiology & Perioperative Medicine, Portland, OR, USA
| | - Meike J Saul
- Technische Universität Darmstadt, Department of Biology, Darmstadt, Germany
| | - Raymond M Schiffelers
- University Medical Center Utrecht, Laboratory for Clinical Chemistry & Hematology, Utrecht, The Netherlands
| | - Raphael Schneider
- University of Toronto, Department of Laboratory Medicine and Pathobiology, Toronto, Canada
- University of Toronto, Department of Medicine, Division of Neurology, Toronto, Canada
| | - Tine Hiorth Schøyen
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | | | - Eriomina Shahaj
- Fondazione IRCCS Istituto Nazionale dei Tumori, Unit of Immunotherapy of Human Tumors, Milan, Italy
| | - Shivani Sharma
- University of California, Los Angeles, California NanoSystems Institute, Los Angeles, CA, USA
- University of California, Los Angeles, Department of Pathology and Laboratory Medicine, Los Angeles, CA, USA
- University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Olga Shatnyeva
- AstraZeneca, Discovery Sciences, IMED Biotech Unit, Gothenburg, Sweden
| | - Faezeh Shekari
- Royan Institute for Stem Cell Biology and Technology, ACECR, Cell Science Research Center, Department of Stem Cells and Developmental Biology, Tehran, Iran
| | - Ganesh Vilas Shelke
- University of Gothenburg, Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Cancer Center, Gothenburg, Sweden
- University of Gothenburg, Institute of Medicine at Sahlgrenska Academy, Krefting Research Centre, Gothenburg, Sweden
| | - Ashok K Shetty
- Research Service, Olin E. Teague Veterans’ Medical Center, Temple, TX, USA
- Texas A&M University College of Medicine, Institute for Regenerative Medicine and Department of Molecular and Cellular Medicine, College Station, TX, USA
| | | | - Pia R-M Siljander
- University of Helsinki, EV Core Facility, Helsinki, Finland
- University of Helsinki, Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Programme, EV group, Helsinki, Finland
| | - Andreia M Silva
- INEB - Instituto de Engenharia Biomédica, Porto, Portugal
- University of Porto, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- University of Porto, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Porto, Portugal
| | - Agata Skowronek
- Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Orman L Snyder
- Kansas State University, College of Veterinary Medicine, Manhattan, KS, USA
| | | | - Barbara W Sódar
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - Carolina Soekmadji
- QIMR Berghofer Medical Research Institute, Herston, Australia
- The University of Queensland, Brisbane, Australia
| | - Javier Sotillo
- James Cook University, Australian Institute of Tropical Health and Medicine, Centre for Biodiscovery and Molecular Development of Therapeutics, Cairns, Australia
| | | | - Willem Stoorvogel
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | - Shannon L Stott
- Harvard Medical School, Department of Medicine, Boston, MA, USA
- Massachusetts General Cancer Center, Boston, MA, USA
| | - Erwin F Strasser
- FAU Erlangen-Nuremberg, Transfusion and Haemostaseology Department, Erlangen, Germany
| | - Simon Swift
- University of Auckland, Department of Molecular Medicine and Pathology, Auckland, New Zealand
| | - Hidetoshi Tahara
- Hiroshima University, Institute of Biomedical & Health Sciences, Department of Cellular and Molecular Biology, Hiroshima, Japan
| | - Muneesh Tewari
- University of Michigan, Biointerfaces Institute, Ann Arbor, MI, USA
- University of Michigan, Department of Biomedical Engineering, Ann Arbor, MI, USA
- University of Michigan, Department of Internal Medicine - Hematology/Oncology Division, Ann Arbor, MI, USA
| | - Kate Timms
- University of Manchester, Manchester, UK
| | - Swasti Tiwari
- Georgetown University, Department of Medicine, Washington, DC, USA
- Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Molecular Medicine & Biotechnology, Lucknow, India
| | - Rochelle Tixeira
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Mercedes Tkach
- Institut Curie, INSERM U932, PSL Research University, Paris, France
| | - Wei Seong Toh
- National University of Singapore, Faculty of Dentistry, Singapore
| | - Richard Tomasini
- INSERM U1068, Aix Marseille University, CNRS UMR7258, Marseille, France
| | | | - Juan Pablo Tosar
- Institut Pasteur de Montevideo, Functional Genomics Unit, Montevideo, Uruguay
- Universidad de la República, Faculty of Science, Nuclear Research Center, Analytical Biochemistry Unit, Montevideo, Uruguay
| | | | - Lorena Urbanelli
- University of Perugia, Department of Chemistry, Biology and Biotechnology, Perugia, Italy
| | - Pieter Vader
- University Medical Center Utrecht, Laboratory for Clinical Chemistry & Hematology, Utrecht, The Netherlands
| | - Bas WM van Balkom
- University Medical Center Utrecht, Department of Nephrology and Hypertension, Utrecht, The Netherlands
| | - Susanne G van der Grein
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | - Jan Van Deun
- Cancer Research Institute Ghent, Ghent, Belgium
- Ghent University, Department of Radiation Oncology and Experimental Cancer Research, Laboratory of Experimental Cancer Research, Ghent, Belgium
| | - Martijn JC van Herwijnen
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | | | | | - Martin E van Royen
- Department of Pathology, Erasmus MC, Erasmus Optical Imaging Centre, Rotterdam, The Netherlands
| | | | - M Helena Vasconcelos
- IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- University of Porto, Faculty of Pharmacy (FFUP), Porto, Portugal
- University of Porto, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
| | - Ivan J Vechetti
- University of Kentucky, College of Medicine, Department of Physiology, Lexington, KY, USA
| | - Tiago D Veit
- Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Departamento de Microbiologia, Imunologia e Parasitologia, Porto Alegre, Brazil
| | - Laura J Vella
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
- The University of Melbourne, The Department of Medicine, Melbourne, Australia
| | - Émilie Velot
- UMR 7365 CNRS-Université de Lorraine, Vandœuvre-lès-Nancy, France
| | | | - Beate Vestad
- Oslo University Hospital Rikshospitalet, Research Institute of Internal Medicine, Oslo, Norway
- Regional Research Network on Extracellular Vesicles, RRNEV, Oslo, Norway
- University of Oslo, Institute of Clinical Medicine, Oslo, Norway
| | - Jose L Viñas
- Kidney Research Centre, Ottawa, Canada
- Ottawa Hospital Research Institute, Ottawa, Canada
- University of Ottawa, Ottawa, Canada
| | - Tamás Visnovitz
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - Krisztina V Vukman
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - Jessica Wahlgren
- University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal, Sweden
| | - Dionysios C Watson
- Case Western Reserve University, Department of Medicine, Cleveland, OH, USA
- University Hospitals Cleveland Medical Center, Department of Medicine, Cleveland, OH, USA
| | - Marca HM Wauben
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | - Alissa Weaver
- Vanderbilt University School of Medicine, Department of Cell and Developmental Biology, Nashville, TN, USA
| | | | - Viktoria Weber
- Danube University Krems, Department for Biomedical Research and Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Krems an der Donau, Austria
| | - Ann M Wehman
- University of Würzburg, Rudolf Virchow Center, Würzburg, Germany
| | - Daniel J Weiss
- The University of Vermont Medical Center, Department of Medicine, Burlington, VT, USA
| | - Joshua A Welsh
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, MD, USA
| | - Sebastian Wendt
- University Hospital RWTH Aachen, Department of Thoracic and Cardiovascular Surgery, Aachen, Germany
| | - Asa M Wheelock
- Karolinska Institute, Department of Medicine and Center for Molecular Medicine, Respiratory Medicine Unit, Stockholm, Sweden
| | - Zoltán Wiener
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - Leonie Witte
- University Medical Center Göttingen, Developmental Biochemistry, Göttingen, Germany
- University Medical Center Göttingen, Hematology and Oncology, Göttingen, Germany
| | - Joy Wolfram
- Chinese Academy of Sciences, Wenzhou Institute of Biomaterials and Engineering, Wenzhou, China
- Houston Methodist Research Institute, Department of Nanomedicine, Houston, TX, USA
- Mayo Clinic, Department of Transplantation Medicine/Department of Physiology and Biomedical Engineering, Jacksonville, FL, USA
| | - Angeliki Xagorari
- George Papanicolaou Hospital, Public Cord Blood Bank, Department of Haematology - BMT Unit, Thessaloniki, Greece
| | - Patricia Xander
- Universidade Federal de São Paulo Campus Diadema, Departamento de Ciências Farmacêuticas, Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, São Paulo, Brazil
| | - Jing Xu
- BC Cancer, Canada’s Michael Smith Genome Sciences Centre, Vancouver, Canada
- Simon Fraser University, Department of Molecular Biology and Biochemistry, Burnaby, Canada
| | - Xiaomei Yan
- Xiamen University, Department of Chemical Biology, Xiamen, China
| | - María Yáñez-Mó
- Centro de Biología Molecular Severo Ochoa, Instituto de Investigación Sanitaria la Princesa (IIS-IP), Madrid, Spain
- Universidad Autónoma de Madrid, Departamento de Biología Molecular, Madrid, Spain
| | - Hang Yin
- Tsinghua University, School of Pharmaceutical Sciences, Beijing, China
| | - Yuana Yuana
- Technical University Eindhoven, Faculty Biomedical Technology, Eindhoven, The Netherlands
| | - Valentina Zappulli
- University of Padova, Department of Comparative Biomedicine and Food Science, Padova, Italy
| | - Jana Zarubova
- Institute of Physiology CAS, Department of Biomaterials and Tissue Engineering, BIOCEV, Vestec, Czech Republic
- Institute of Physiology CAS, Department of Biomaterials and Tissue Engineering, Prague, Czech Republic
- University of California, Los Angeles, Department of Bioengineering, Los Angeles, CA, USA
| | - Vytautas Žėkas
- Vilnius University, Institute of Biomedical Sciences, Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Vilnius, Lithuania
| | - Jian-ye Zhang
- Guangzhou Medical University, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology, Guangzhou, China
| | - Zezhou Zhao
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | - Lei Zheng
- Nanfang Hospital, Southern Medical University, Department of Clinical Laboratory Medicine, Guangzhou, China
| | | | - Antje M Zickler
- Karolinska Institute, Clinical Research Center, Unit for Molecular Cell and Gene Therapy Science, Stockholm, Sweden
| | - Pascale Zimmermann
- Aix-Marseille Université, Institut Paoli-Calmettes, INSERM U1068, CNRS UMR7258, Centre de Recherche en Cancérologie de Marseille, Marseille, France
- KU Leuven (Leuven University), Department of Human Genetics, Leuven, Belgium
| | - Angela M Zivkovic
- University of California, Davis, Department of Nutrition, Davis, CA, USA
| | | | - Ewa K Zuba-Surma
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Kraków, Poland
| |
Collapse
|
45
|
Hadley EE, Sheller-Miller S, Saade G, Salomon C, Mesiano S, Taylor RN, Taylor BD, Menon R. Amnion epithelial cell-derived exosomes induce inflammatory changes in uterine cells. Am J Obstet Gynecol 2018; 219:478.e1-478.e21. [PMID: 30138617 PMCID: PMC6239974 DOI: 10.1016/j.ajog.2018.08.021] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 08/08/2018] [Accepted: 08/13/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Fetal endocrine signals are generally considered to contribute to the timing of birth and the initiation of labor. Fetal tissues under oxidative stress release inflammatory mediators that lead to sterile inflammation within the maternal-fetal interface. Importantly, these inflammatory mediators are packaged into exosomes, bioactive cell-derived extra cellular vesicles that function as vectors and transport them from the fetal side to the uterine tissues where they deposit their cargo into target cells enhancing uterine inflammatory load. This exosome-mediated signaling is a novel mechanism for fetal-maternal communication. OBJECTIVE This report tested the hypothesis that oxidative stress can induce fetal amnion cells to produce exosomes, which function as a paracrine intermediary between the fetus and mother and biochemically signal readiness for parturition. STUDY DESIGN Primary amnion epithelial cells were grown in normal cell culture (control) or exposed to oxidative stress conditions (induced by cigarette smoke extract). Exosomes were isolated from cell supernatant by sequential ultracentrifugation. Exosomes were quantified and characterized based on size, shape, and biochemical markers. Myometrial, decidual, and placental cells (BeWo) were treated with 2 × 105, 2 × 107, and 2 × 109 control or oxidative stress-derived amnion epithelial cell exosomes for 24 hours. Entry of amnion epithelial cell exosomes into cells was confirmed by confocal microscopy of fluorescent-labeled exosomes. The effect of amnion epithelial cell exosomes on target cell inflammatory status was determined by measuring production of interleukin-6, interleukin-8, interleukin-1β, tumor necrosis factor-α, and prostaglandin E2 by enzyme-linked immunosorbent assay and inflammatory gene transcription factor (nuclear factor-κβ) activation status by immunoblotting for phosphorylated RelA/p65. Localization of NANOG in term human myometrium and decidua obtained from women before labor and during labor was performed using immunohistochemistry. Data were analyzed by Wilcoxon-Mann-Whitney test to compare effects of exosomes from control and oxidative stress-treated amnion epithelial cells on inflammatory status of target cells. RESULTS Amnion epithelial cells released ∼125 nm, cup-shaped exosomes with ∼899 and 1211 exosomes released per cell from control and oxidative stress-induced cells, respectively. Amnion epithelial cell exosomes were detected in each target cell type after treatment using confocal microscopy. Treatment with amnion epithelial cell exosomes increased secretion of interleukin-6, interleukin-8, and PGE2 and activation of NF-κβ (each P < .05) in myometrial and decidual cells. Exosome treatments had no effect on interleukin-6 and PGE2 production in BeWo cells. NANOG staining was higher in term labor myometrium and decidua compared to tissues not in labor. CONCLUSION In vitro, amnion epithelial cell exosomes lead to an increased inflammatory response in maternal uterine cells whereas placental cells showed refractoriness. Fetal cell exosomes may function to signal parturition by increasing maternal gestational cell inflammation.
Collapse
Affiliation(s)
- Emily E Hadley
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX
| | - Samantha Sheller-Miller
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX; Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX
| | - George Saade
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX
| | - Carlos Salomon
- Exosome Biology Laboratory, Center for Clinical Diagnostics, Center for Clinical Research, Royal Brisbane and Women's Hospital, University of Queensland, Brisbane, Australia; Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepción, Concepción, Chile
| | - Sam Mesiano
- Department of Reproductive Biology, Case Western Reserve University, School of Medicine, Cleveland, OH
| | - Robert N Taylor
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, NC
| | - Brandie D Taylor
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, TX
| | - Ramkumar Menon
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX.
| |
Collapse
|
46
|
Bijnsdorp IV, van Royen ME, Verhaegh GW, Martens-Uzunova ES. The Non-Coding Transcriptome of Prostate Cancer: Implications for Clinical Practice. Mol Diagn Ther 2018; 21:385-400. [PMID: 28299719 PMCID: PMC5511609 DOI: 10.1007/s40291-017-0271-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prostate cancer (PCa) is the most common type of cancer and the second leading cause of cancer-related death in men. Despite extensive research, the molecular mechanisms underlying PCa initiation and progression remain unclear, and there is increasing need of better biomarkers that can distinguish indolent from aggressive and life-threatening disease. With the advent of advanced genomic technologies in the last decade, it became apparent that the human genome encodes tens of thousands non-protein-coding RNAs (ncRNAs) with yet to be discovered function. It is clear now that the majority of ncRNAs exhibit highly specific expression patterns restricted to certain tissues and organs or developmental stages and that the expression of many ncRNAs is altered in disease and cancer, including cancer of the prostate. Such ncRNAs can serve as important biomarkers for PCa diagnosis, prognosis, or prediction of therapy response. In this review, we give an overview of the different types of ncRNAs and their function, describe ncRNAs relevant for the diagnosis and prognosis of PCa, and present emerging new aspects of ncRNA research that may contribute to the future utilization of ncRNAs as clinically useful therapeutic targets.
Collapse
MESH Headings
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/urine
- Early Detection of Cancer/methods
- Gene Expression Regulation, Neoplastic
- High-Throughput Nucleotide Sequencing
- Humans
- Male
- Molecular Targeted Therapy
- Precision Medicine
- Prognosis
- Prostatic Neoplasms/diagnosis
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- RNA, Untranslated/blood
- RNA, Untranslated/classification
- RNA, Untranslated/genetics
- RNA, Untranslated/urine
Collapse
Affiliation(s)
- Irene V Bijnsdorp
- Department of Urology, VU University Medical Center, Amsterdam, The Netherlands
| | - Martin E van Royen
- Department of Pathology and Erasmus Optical Imaging Centre (OIC), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Gerald W Verhaegh
- Department of Urology, Radboud university medical center, Nijmegen, The Netherlands
| | - Elena S Martens-Uzunova
- Department of Urology, Erasmus Medical Center, Erasmus Cancer Institute, Room Be-362b, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
47
|
Epigenetic modifications in hyperhomocysteinemia: potential role in diabetic retinopathy and age-related macular degeneration. Oncotarget 2018; 9:12562-12590. [PMID: 29560091 PMCID: PMC5849155 DOI: 10.18632/oncotarget.24333] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 01/24/2018] [Indexed: 02/03/2023] Open
Abstract
To study Hyperhomocysteinemia (HHcy)-induced epigenetic modifications as potential mechanisms of blood retinal barrier (BRB) dysfunction, retinas isolated from three- week-old mice with elevated level of Homocysteine (Hcy) due to lack of the enzyme cystathionine β-synthase (cbs-/- , cbs+/- and cbs+/+ ), human retinal endothelial cells (HRECs), and human retinal pigmented epithelial cells (ARPE-19) treated with or without Hcy were evaluated for (1) histone deacetylases (HDAC), (2) DNA methylation (DNMT), and (3) miRNA analysis. Differentially expressed miRNAs in mice with HHcy were further compared with miRNA analysis of diabetic mice retinas (STZ) and miRNAs within the exosomes released from Hcy-treated RPEs. Differentially expressed miRNAs were further evaluated for predicted target genes and associated pathways using Ingenuity Pathway Analysis. HHcy significantly increased HDAC and DNMT activity in HRECs, ARPE-19, and cbs mice retinas, whereas inhibition of HDAC and DNMT decreased Hcy-induced BRB dysfunction. MiRNA profiling detected 127 miRNAs in cbs+/- and 39 miRNAs in cbs-/- mice retinas, which were significantly differentially expressed compared to cbs+/+ . MiRNA pathway analysis showed their involvement in HDAC and DNMT activation, endoplasmic reticulum (ER), and oxidative stresses, inflammation, hypoxia, and angiogenesis pathways. Hcy-induced epigenetic modifications may be involved in retinopathies associated with HHcy, such as age-related macular degeneration and diabetic retinopathy.
Collapse
|
48
|
Diet-resistant obesity is characterized by a distinct plasma proteomic signature and impaired muscle fiber metabolism. Int J Obes (Lond) 2017; 42:353-362. [PMID: 29151592 PMCID: PMC5880582 DOI: 10.1038/ijo.2017.286] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/11/2017] [Accepted: 10/30/2017] [Indexed: 12/28/2022]
Abstract
Background/Objectives: Inter-individual variability in weight loss during obesity treatment is complex and poorly understood. Here we use whole body and tissue approaches to investigate fuel oxidation characteristics in skeletal muscle fibers, cells and distinct circulating protein biomarkers before and after a high fat meal (HFM) challenge in those who lost the most (obese diet-sensitive; ODS) vs the least (obese diet-resistant; ODR) amount of weight in a highly controlled weight management program. Subjects/Methods: In 20 weight stable-matched ODS and ODR women who previously completed a standardized clinical weight loss program, we analyzed whole-body energetics and metabolic parameters in vastus lateralis biopsies and plasma samples that were obtained in the fasting state and 6 h after a defined HFM, equivalent to 35% of total daily energy requirements. Results: At baseline (fasting) and post-HFM, muscle fatty acid oxidation and maximal oxidative phosphorylation were significantly greater in ODS vs ODR, as was reactive oxygen species emission. Plasma proteomics of 1130 proteins pre and 1, 2, 5 and 6 h after the HFM demonstrated distinct group and interaction differences. Group differences identified S-formyl glutathione hydratase, heat shock 70 kDA protein 1A/B (HSP72), and eukaryotic translation initiation factor 5 (eIF5) to be higher in ODS vs ODR. Group-time differences included aryl hydrocarbon interacting protein (AIP), peptidylpropyl isomerase D (PPID) and tyrosine protein-kinase Fgr, which increased in ODR vs ODS over time. HSP72 levels correlated with muscle oxidation and citrate synthase activity. These proteins circulate in exosomes; exosomes isolated from ODS plasma increased resting, leak and maximal respiration rates in C2C12 myotubes by 58%, 21% and 51%, respectively, vs those isolated from ODR plasma. Conclusions: Findings demonstrate distinct muscle metabolism and plasma proteomics in fasting and post-HFM states corresponding in diet-sensitive vs diet-resistant obese women.
Collapse
|
49
|
Thayanithy V, O'Hare P, Wong P, Zhao X, Steer CJ, Subramanian S, Lou E. A transwell assay that excludes exosomes for assessment of tunneling nanotube-mediated intercellular communication. Cell Commun Signal 2017; 15:46. [PMID: 29132390 PMCID: PMC5683209 DOI: 10.1186/s12964-017-0201-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 10/30/2017] [Indexed: 12/25/2022] Open
Abstract
Background Tunneling nanotubes (TNTs) are naturally-occurring filamentous actin-based membranous extensions that form across a wide spectrum of mammalian cell types to facilitate long-range intercellular communication. Valid assays are needed to accurately assess the downstream effects of TNT-mediated transfer of cellular signals in vitro. We recently reported a modified transwell assay system designed to test the effects of intercellular transfer of a therapeutic oncolytic virus, and viral-activated drugs, between cells via TNTs. The objective of the current study was to demonstrate validation of this in vitro approach as a new method for effectively excluding diffusible forms of long- and close-range intercellular transfer of intracytoplasmic cargo, including exosomes/microvesicles and gap junctions in order to isolate TNT-selective cell communication. Methods We designed several steps to effectively reduce or eliminate diffusion and long-range transfer via these extracellular vesicles, and used Nanoparticle Tracking Analysis to quantify exosomes following implementation of these steps. Results The experimental approach outlined here effectively reduced exosome trafficking by >95%; further use of heparin to block exosome uptake by putative recipient cells further impeded transfer of these extracellular vesicles. Conclusions This validated assay incorporates several steps that can be taken to quantifiably control for extracellular vesicles in order to perform studies focused on TNT-selective communication. Electronic supplementary material The online version of this article (10.1186/s12964-017-0201-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Venugopal Thayanithy
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Mayo Mail Code 480, 420 Delaware Street SE, Minneapolis, MN, 55455, USA.,Present Address: Molecular Diagnostics Laboratory, University of Minnesota Medical Center, Fairview, 420 Delaware St SE, MMC 198, Minneapolis, MN, 55455, USA
| | - Patrick O'Hare
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Mayo Mail Code 480, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Phillip Wong
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Mayo Mail Code 480, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Xianda Zhao
- Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Clifford J Steer
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota, Minneapolis, MN, 55455, USA.,Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, 55455, USA
| | | | - Emil Lou
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Mayo Mail Code 480, 420 Delaware Street SE, Minneapolis, MN, 55455, USA. .,Graduate Faculty, Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
50
|
Neutral Sphingomyelinase-2 Deficiency Ameliorates Alzheimer's Disease Pathology and Improves Cognition in the 5XFAD Mouse. J Neurosci 2017; 36:8653-67. [PMID: 27535912 DOI: 10.1523/jneurosci.1429-16.2016] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 06/27/2016] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED Recent evidence implicates exosomes in the aggregation of Aβ and spreading of tau in Alzheimer's disease. In neural cells, exosome formation can be blocked by inhibition or silencing of neutral sphingomyelinase-2 (nSMase2). We generated genetically nSMase2-deficient 5XFAD mice (fro;5XFAD) to assess AD-related pathology in a mouse model with consistently reduced ceramide generation. We conducted in vitro assays to assess Aβ42 aggregation and glial clearance with and without exosomes isolated by ultracentrifugation and determined exosome-induced amyloid aggregation by particle counting. We analyzed brain exosome content, amyloid plaque formation, neuronal degeneration, sphingolipid, Aβ42 and phospho-tau levels, and memory-related behaviors in 5XFAD versus fro;5XFAD mice using contextual and cued fear conditioning. Astrocyte-derived exosomes accelerated aggregation of Aβ42 and blocked glial clearance of Aβ42 in vitro Aβ42 aggregates were colocalized with extracellular ceramide in vitro using a bifunctional ceramide analog preloaded into exosomes and in vivo using anticeramide IgG, implicating ceramide-enriched exosomes in plaque formation. Compared with 5XFAD mice, the fro;5XFAD mice had reduced brain exosomes, ceramide levels, serum anticeramide IgG, glial activation, total Aβ42 and plaque burden, tau phosphorylation, and improved cognition in a fear-conditioned learning task. Ceramide-enriched exosomes appear to exacerbate AD-related brain pathology by promoting the aggregation of Aβ. Reduction of exosome secretion by nSMase2 loss of function improves pathology and cognition in the 5XFAD mouse model. SIGNIFICANCE STATEMENT We present for the first time evidence, using Alzheimer's disease (AD) model mice deficient in neural exosome secretion due to lack of neutral sphingomyelinase-2 function, that ceramide-enriched exosomes exacerbate AD-related pathologies and cognitive deficits. Our results provide rationale to pursue a means of inhibiting exosome secretion as a potential therapy for individuals at risk for developing AD.
Collapse
|