Jae Hoon Sul, Sol Shin, Hark Kyun Kim, Jihoon Han, Junsik Kim, Soyong Son, Jungmi Lee, Seung Hyun Baek, Yoonsuk Cho, Jeongmi Lee, Jinsu Park, Donghoon Ahn, Sunyoung Park, Leon F. Palomera, Jeein Lim, Jongho Kim, Chanhee Kim, Seungsu Han, Ka Young Chung, Sangho Lee, Tae-in Kam, Yunjong Lee, Jeongyun Kim, Jae Hyung Park, Dong-Gyu Jo
{"title":"Dopamine-conjugated extracellular vesicles induce autophagy in Parkinson's disease","authors":"Jae Hoon Sul, Sol Shin, Hark Kyun Kim, Jihoon Han, Junsik Kim, Soyong Son, Jungmi Lee, Seung Hyun Baek, Yoonsuk Cho, Jeongmi Lee, Jinsu Park, Donghoon Ahn, Sunyoung Park, Leon F. Palomera, Jeein Lim, Jongho Kim, Chanhee Kim, Seungsu Han, Ka Young Chung, Sangho Lee, Tae-in Kam, Yunjong Lee, Jeongyun Kim, Jae Hyung Park, Dong-Gyu Jo","doi":"10.1002/jev2.70018","DOIUrl":"10.1002/jev2.70018","url":null,"abstract":"<p>The application of extracellular vesicles (EVs) as vehicles for anti-Parkinson's agents represents a significant advance, yet their clinical translation is hampered by challenges in efficient brain delivery and complex blood-brain barrier (BBB) targeting strategies. In this study, we engineered dopamine onto the surface of adipose-derived stem cell EVs (Dopa-EVs) utilizing a facile, two-step cross-linking approach. This engineering enhanced neuronal uptake of the EVs in primary neurons and neuroblastoma cells, a process shown to be competitively inhibited by dopamine pretreatment and dopamine receptor antibodies. Notably, Dopa-EVs demonstrated increased brain accumulation in mouse Parkinson's disease (PD) models. Therapeutically, Dopa-EVs administration led to the rescue of dopaminergic neuronal loss and amelioration of behavioural deficits in both 6-hydroxydopamine (6-OHDA) and α-Syn PFF-induced PD models. Furthermore, we observed that Dopa-EVs stimulated autophagy evidenced by the upregulation of Beclin-1 and LC3-II. These findings collectively indicate that surface modification of EVs with dopamine presents a potent strategy for targeting dopaminergic neurons in the brain. The remarkable therapeutic potential of Dopa-EVs, demonstrated in PD models, positions them as a highly promising candidate for PD treatment, offering a significant advance over current therapeutic modalities.</p>","PeriodicalId":15811,"journal":{"name":"Journal of Extracellular Vesicles","volume":"13 12","pages":""},"PeriodicalIF":15.5,"publicationDate":"2024-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11621972/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142785505","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Rebecca T. Miceli, Tzu-Yi Chen, Yohei Nose, Swapnil Tichkule, Briana Brown, John F. Fullard, Marilyn D. Saulsbury, Simon O. Heyliger, Sacha Gnjatic, Natasha Kyprianou, Carlos Cordon-Cardo, Susmita Sahoo, Emanuela Taioli, Panos Roussos, Gustavo Stolovitzky, Edgar Gonzalez-Kozlova, Navneet Dogra
{"title":"Extracellular vesicles, RNA sequencing, and bioinformatic analyses: Challenges, solutions, and recommendations","authors":"Rebecca T. Miceli, Tzu-Yi Chen, Yohei Nose, Swapnil Tichkule, Briana Brown, John F. Fullard, Marilyn D. Saulsbury, Simon O. Heyliger, Sacha Gnjatic, Natasha Kyprianou, Carlos Cordon-Cardo, Susmita Sahoo, Emanuela Taioli, Panos Roussos, Gustavo Stolovitzky, Edgar Gonzalez-Kozlova, Navneet Dogra","doi":"10.1002/jev2.70005","DOIUrl":"https://doi.org/10.1002/jev2.70005","url":null,"abstract":"<p>Extracellular vesicles (EVs) are heterogeneous entities secreted by cells into their microenvironment and systemic circulation. Circulating EVs carry functional small RNAs and other molecular footprints from their cell of origin, and thus have evident applications in liquid biopsy, therapeutics, and intercellular communication. Yet, the complete transcriptomic landscape of EVs is poorly characterized due to critical limitations including variable protocols used for EV-RNA extraction, quality control, cDNA library preparation, sequencing technologies, and bioinformatic analyses. Consequently, there is a gap in knowledge and the need for a standardized approach in delineating EV-RNAs. Here, we address these gaps by describing the following points by (1) focusing on the large canopy of the EVs and particles (EVPs), which includes, but not limited to – exosomes and other large and small EVs, lipoproteins, exomeres/supermeres, mitochondrial-derived vesicles, RNA binding proteins, and cell-free DNA/RNA/proteins; (2) examining the potential functional roles and biogenesis of EVPs; (3) discussing various transcriptomic methods and technologies used in uncovering the cargoes of EVPs; (4) presenting a comprehensive list of RNA subtypes reported in EVPs; (5) describing different EV-RNA databases and resources specific to EV-RNA species; (6) reviewing established bioinformatics pipelines and novel strategies for reproducible EV transcriptomics analyses; (7) emphasizing the significant need for a gold standard approach in identifying EV-RNAs across studies; (8) and finally, we highlight current challenges, discuss possible solutions, and present recommendations for robust and reproducible analyses of EVP-associated small RNAs. Overall, we seek to provide clarity on the transcriptomics landscape, sequencing technologies, and bioinformatic analyses of EVP-RNAs. Detailed portrayal of the current state of EVP transcriptomics will lead to a better understanding of how the RNA cargo of EVPs can be used in modern and targeted diagnostics and therapeutics. For the inclusion of different particles discussed in this article, we use the terms large/small EVs, non-vesicular extracellular particles (NVEPs), EPs and EVPs as defined in MISEV guidelines by the International Society of Extracellular Vesicles (ISEV).</p>","PeriodicalId":15811,"journal":{"name":"Journal of Extracellular Vesicles","volume":"13 12","pages":""},"PeriodicalIF":15.5,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/jev2.70005","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142764330","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ephraim A. Ansa-Addo, Paras Pathak, Maria V. McCrossan, Izadora Volpato Rossi, Mahamed Abdullahi, Dan Stratton, Sigrun Lange, Marcel I. Ramirez, Jameel M. Inal
{"title":"Monocyte-derived extracellular vesicles, stimulated by Trypanosoma cruzi, enhance cellular invasion in vitro via activated TGF-β1","authors":"Ephraim A. Ansa-Addo, Paras Pathak, Maria V. McCrossan, Izadora Volpato Rossi, Mahamed Abdullahi, Dan Stratton, Sigrun Lange, Marcel I. Ramirez, Jameel M. Inal","doi":"10.1002/jev2.70014","DOIUrl":"https://doi.org/10.1002/jev2.70014","url":null,"abstract":"<p>During cell invasion, large Extracellular Vesicle (lEV) release from host cells was dose-dependently triggered by <i>Trypanosoma cruzi</i> metacyclic trypomastigotes (Mtr). This lEV release was inhibited when IP<sub>3</sub>-mediated Ca<sup>2+</sup> exit from the ER and further Ca<sup>2+</sup> entry from plasma membrane channels was blocked, but whilst any store-independent Ca<sup>2+</sup> entry (SICE) could continue unabated. That lEV release was equally inhibited if all entry from external sources was blocked by chelation of external Ca<sup>2+</sup> points to the major contributor to Mtr-triggered host cell lEV release being IP<sub>3</sub>/store-mediated Ca<sup>2+</sup> release, SICE playing a minor role. Host cell lEVs were released through Mtr interaction with host cell lipid raft domains, integrins, and mechanosensitive ion channels, whereupon [Ca<sup>2+</sup>]<sub>cyt</sub> increased (50 to 750 nM) within 15 s. lEV release and cell entry of <i>T. cruzi</i>, which increased up to 30 and 60 mpi, respectively, as well as raised actin depolymerization at 60 mpi, were all reduced by TRPC inhibitor, GsMTx-4. Vesicle release and infection was also reduced with RGD peptide, methyl-β-cyclodextrin, knockdown of calpain and with the calpain inhibitor, calpeptin. Restoration of lEV levels, whether with lEVs from infected or uninfected epithelial cells, did not restore invasion, but supplementation with lEVs from infected monocytes, did. We provide evidence of THP-1 monocyte-derived lEV interaction with Mtr (lipid mixing by R18-dequenching; flow cytometry showing transfer to Mtr of R18 from R18-lEVs and of LAP(TGF-β1). Active, mature TGF-β1 (at 175 pg/×10<sup>5</sup> in THP-1 lEVs) was detected in concentrated lEV-/cell-free supernatant by western blotting, only after THP-1 lEVs had interacted with Mtr. The TGF-β1 receptor (TβRI) inhibitor, SB-431542, reduced the enhanced cellular invasion due to monocyte-lEVs.</p>","PeriodicalId":15811,"journal":{"name":"Journal of Extracellular Vesicles","volume":"13 11","pages":""},"PeriodicalIF":15.5,"publicationDate":"2024-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/jev2.70014","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142749253","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Kewei Wang, Qin He, Mengmeng Yang, Qincheng Qiao, Jun Chen, Jia Song, Nan Zang, Huiqing Hu, Longqing Xia, Yingyue Xiang, Fei Yan, Xinguo Hou, Li Chen
{"title":"Glycoengineered extracellular vesicles released from antibacterial hydrogel facilitate diabetic wound healing by promoting angiogenesis","authors":"Kewei Wang, Qin He, Mengmeng Yang, Qincheng Qiao, Jun Chen, Jia Song, Nan Zang, Huiqing Hu, Longqing Xia, Yingyue Xiang, Fei Yan, Xinguo Hou, Li Chen","doi":"10.1002/jev2.70013","DOIUrl":"10.1002/jev2.70013","url":null,"abstract":"<p>Diabetic wounds have become a global healthcare burden owing to impaired angiogenesis and persistent infections. Extracellular vesicles (EVs) can improve diabetic wounds, though their targeting ability is limited. In this study, we investigated the performance of a novel hydrogel dressing comprised of gelatin methacryloyl, glycoengineered EVs, and polylysine in treating infected diabetic wounds. High-throughput single-cell RNA sequencing (scRNA-seq) and immunofluorescence staining revealed that E-selectin (SELE) levels were higher in diabetic wounds than in non-diabetic wounds. Mesenchymal stromal cells (MSCs) were transfected with a lentivirus containing fucosyltransferase VII (FUT7) and a CD63-P19-Nluc vector to enhance the expression of sialyl Lewis X (sLeX), the ligand of E-selectin, on the surface of EVs (s-EVs) derived from transfected MSCs (s-MSCs). s-EVs can target human umbilical vein endothelial cells (HUVECs) under lipopolysaccharide stimulation and promote the function of stimulated HUVECs in vitro. To promote and sustain the release of s-EVs, we fabricated a gelatin methacryloyl (Gel)/poly-L-lysine methacryloyl (PL)-5 hydrogel with good antibacterial ability, biocompatibility and mechanical properties. In a mouse experiment, s-EV@Gel/PL-5 exhibited excellent angiogenesis and anti-inflammatory abilities and further promoted the healing of infected diabetic wounds. Our findings demonstrated the potential of the s-EV@Gel/PL-5 hydrogel in the clinical treatment of diabetic infectious wounds.</p>","PeriodicalId":15811,"journal":{"name":"Journal of Extracellular Vesicles","volume":"13 11","pages":""},"PeriodicalIF":15.5,"publicationDate":"2024-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/jev2.70013","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142729525","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Diego de Miguel-Perez, Marisol Arroyo-Hernandez, Sabrina La Salvia, Muthukumar Gunasekaran, Edward M. Pickering, Stephanie Avila, Etse Gebru, Eduardo Becerril-Vargas, Sergio Monraz-Perez, Kapil Saharia, Alison Grazioli, Michael T. McCurdy, Matthew Frieman, Lisa Miorin, Alessandro Russo, Andrés F. Cardona, Adolfo García-Sastre, Sunjay Kaushal, Fred R. Hirsch, Djordje Atanackovic, Susmita Sahoo, Oscar Arrieta, Christian Rolfo
{"title":"Extracellular vesicles containing SARS-CoV-2 proteins are associated with multi-organ dysfunction and worse outcomes in patients with severe COVID-19","authors":"Diego de Miguel-Perez, Marisol Arroyo-Hernandez, Sabrina La Salvia, Muthukumar Gunasekaran, Edward M. Pickering, Stephanie Avila, Etse Gebru, Eduardo Becerril-Vargas, Sergio Monraz-Perez, Kapil Saharia, Alison Grazioli, Michael T. McCurdy, Matthew Frieman, Lisa Miorin, Alessandro Russo, Andrés F. Cardona, Adolfo García-Sastre, Sunjay Kaushal, Fred R. Hirsch, Djordje Atanackovic, Susmita Sahoo, Oscar Arrieta, Christian Rolfo","doi":"10.1002/jev2.70001","DOIUrl":"https://doi.org/10.1002/jev2.70001","url":null,"abstract":"<p>The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19) and has been related to more than 7 million deaths globally since 2019. The association of high levels of IL-6 with severe cases led to the early evaluation of the anti-IL6 inhibitor tocilizumab as a potential treatment, which unfortunately failed to improve survival in many trials. Moreover, little is known about the development of COVID-19 sequelae, and biomarkers are needed to understand and anticipate these processes. Because extracellular vesicles (EVs) play an important role in viral infection and immune response, they could potentially serve as predictive and prognostic biomarkers. We isolated EVs from 39 patients with severe COVID-19, from which 29 received tocilizumab and 10 were considered controls. Blood samples, which were collected at hospitalisation before treatment, at Day 7, and Day 15 during follow-up, were assessed by immunoblot for longitudinal expression of spike (S) and nucleocapsid (N) proteins. Dynamic expression was calculated and compared with clinicopathological and experimental variables. Expression of EV S was validated by immunogold and imaging flow-cytometry, revealing an enrichment in CD9+ EVs. As a result, decreasing expression of EV viral proteins was observed in patients treated with tocilizumab. Moreover, higher increase in EV S was observed in patients with lower antibody response, hyperfibrinogenemia, lower respiratory function, higher blood pressure and shorter outcomes. These findings lay the foundation for future studies characterizing the role of EVs in multiorgan assessment and identifying biomarkers in patients with severe COVID-19 and possible long COVID.</p>","PeriodicalId":15811,"journal":{"name":"Journal of Extracellular Vesicles","volume":"13 11","pages":""},"PeriodicalIF":15.5,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/jev2.70001","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142666152","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Efficient enzyme-free isolation of brain-derived extracellular vesicles","authors":"Andreu Matamoros-Angles, Emina Karadjuzovic, Behnam Mohammadi, Feizhi Song, Santra Brenna, Susanne Caroline Meister, Bente Siebels, Hannah Voß, Carolin Seuring, Isidre Ferrer, Hartmut Schlüter, Matthias Kneussel, Hermann Clemens Altmeppen, Michaela Schweizer, Berta Puig, Mohsin Shafiq, Markus Glatzel","doi":"10.1002/jev2.70011","DOIUrl":"10.1002/jev2.70011","url":null,"abstract":"<p>Extracellular vesicles (EVs) have gained significant attention as pathology mediators and potential diagnostic tools for neurodegenerative diseases. However, isolation of brain-derived EVs (BDEVs) from tissue remains challenging, often involving enzymatic digestion steps that may compromise the integrity of EV proteins and overall functionality. Here, we describe that collagenase digestion, commonly used for BDEV isolation, produces undesired protein cleavage of EV-associated proteins in brain tissue homogenates and cell-derived EVs. In order to avoid this effect, we studied the possibility of isolating BDEVs with a reduced amount of collagenase or without any protease. Characterization of the isolated BDEVs from mouse and human samples (both female and male) revealed their characteristic morphology and size distribution with both approaches. However, we show that even minor enzymatic digestion induces ‘artificial’ proteolytic processing in key BDEV markers, such as Flotillin-1, CD81, and the cellular prion protein (PrP<sup>C</sup>), whereas avoiding enzymatic treatment completely preserves their integrity. We found no major differences in mRNA and protein content between non-enzymatically and enzymatically isolated BDEVs, suggesting that the same BDEV populations are purified with both approaches. Intriguingly, the lack of Golgi marker GM130 signal, often referred to as contamination indicator (or negative marker) in EV preparations, seems to result from enzymatic digestion rather than from its actual absence in BDEV samples. Overall, we show that non-enzymatic isolation of EVs from brain tissue is possible and avoids artificial pruning of proteins while achieving an overall high BDEV yield and purity. This protocol will help to understand the functions of BDEV and their associated proteins in a near-physiological setting, thus opening new research approaches.</p>","PeriodicalId":15811,"journal":{"name":"Journal of Extracellular Vesicles","volume":"13 11","pages":""},"PeriodicalIF":15.5,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11541858/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142604686","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Margarida Viola, Maarten P. Bebelman, Renee G. C. Maas, Willemijn S. de Voogt, Frederik J. Verweij, Cor S. Seinen, Saskia C. A. de Jager, Pieter Vader, Dirk Michiel Pegtel, Joost Petrus Gerardus Sluijter
{"title":"Hypoxia and TNF-alpha modulate extracellular vesicle release from human induced pluripotent stem cell-derived cardiomyocytes","authors":"Margarida Viola, Maarten P. Bebelman, Renee G. C. Maas, Willemijn S. de Voogt, Frederik J. Verweij, Cor S. Seinen, Saskia C. A. de Jager, Pieter Vader, Dirk Michiel Pegtel, Joost Petrus Gerardus Sluijter","doi":"10.1002/jev2.70000","DOIUrl":"10.1002/jev2.70000","url":null,"abstract":"<p>Extracellular vesicles (EVs) have emerged as important mediators of intercellular communication in the heart under homeostatic and pathological conditions, such as myocardial infarction (MI). However, the basic mechanisms driving cardiomyocyte-derived EV (CM-EV) production following stress are poorly understood. In this study, we generated human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) that express NanoLuc-tetraspanin reporters. These modified hiPSC-CMs allow for quantification of tetraspanin-positive CM-EV secretion from small numbers of cells without the need for time-consuming EV isolation techniques. We subjected these cells to a panel of small molecules to study their effect on CM-EV biogenesis and secretion under basal and stress-associated conditions. We observed that EV biogenesis is context-dependent in hiPSC-CMs. Nutrient starvation decreases CM-EV secretion while hypoxia increases the production of CM-EVs in a nSmase2-dependent manner. Moreover, the inflammatory cytokine TNF-α increased CM-EV secretion through a process involving NLRP3 inflammasome activation and mTOR signalling. Here, we detailed for the first time the regulatory mechanisms of EV biogenesis in hiPSC-CMs upon MI-associated stressors.</p>","PeriodicalId":15811,"journal":{"name":"Journal of Extracellular Vesicles","volume":"13 11","pages":""},"PeriodicalIF":15.5,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11541862/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142604691","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hang Zhao, Zhi Li, Da Liu, Jiaxun Zhang, Zhicheng You, Yuzhang Shao, Hongyan Li, Jun Yang, Xiang Liu, Miaozhu Wang, Chengen Wu, Jing Chen, Jianwu Wang, Guanyi Kong, Libo Zhao
{"title":"PlexinA1 (PLXNA1) as a novel scaffold protein for the engineering of extracellular vesicles","authors":"Hang Zhao, Zhi Li, Da Liu, Jiaxun Zhang, Zhicheng You, Yuzhang Shao, Hongyan Li, Jun Yang, Xiang Liu, Miaozhu Wang, Chengen Wu, Jing Chen, Jianwu Wang, Guanyi Kong, Libo Zhao","doi":"10.1002/jev2.70012","DOIUrl":"10.1002/jev2.70012","url":null,"abstract":"<p>Extracellular vesicles (EVs) had been described as a next-generation drug delivery system, due to the compelling evidence that they can facilitate the transfer of a variety of biomolecules between cells. The most frequently used strategy for loading protein cargoes is the endogenous engineering of EVs through genetic fusion of the protein of interest (POI) and scaffold proteins with high EV-sorting ability. However, the lack of scaffold proteins had become a major issue hindering the promotion of this technology. Herein, we proposed novel screening criteria that relax the inclusion requirement of candidate scaffold proteins and eventually identified a new scaffold protein, PLXNA1. The truncated PLXNA1 not only inherits the high EV-sorting ability of its full-length counterpart but also allows the fusion expression of POI in both outer surface and luminal areas, individually or simultaneously. In conclusion, our screening criteria expanded the range of potential scaffold proteins. The identified scaffold protein PLXNA1 showed great potential in developing therapeutic EVs.</p>","PeriodicalId":15811,"journal":{"name":"Journal of Extracellular Vesicles","volume":"13 11","pages":""},"PeriodicalIF":15.5,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11541859/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142604712","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Emma Tordoff, Jillian Allen, Katya Elgart, Ahmed Elsherbini, Vrinda Kalia, Haotian Wu, Erden Eren, Dimitrios Kapogiannis, Olesia Gololobova, Kenneth Witwer, Olga Volpert, Erez Eitan
{"title":"A novel multiplexed immunoassay for surface-exposed proteins in plasma extracellular vesicles","authors":"Emma Tordoff, Jillian Allen, Katya Elgart, Ahmed Elsherbini, Vrinda Kalia, Haotian Wu, Erden Eren, Dimitrios Kapogiannis, Olesia Gololobova, Kenneth Witwer, Olga Volpert, Erez Eitan","doi":"10.1002/jev2.70007","DOIUrl":"https://doi.org/10.1002/jev2.70007","url":null,"abstract":"<p>Small membranous extracellular vesicles (EV) incorporate proteins and nucleic acids from the parent cell. Proteins exposed on EV surface are dictated by cellular origin and biogenesis pathway. To better understand the EV origin and function, it is important to develop methods that reveal surface protein composition of heterogeneous EV populations in culture supernatants and in biofluids. Tetraspanins CD9, CD63, and CD81 are common and abundant EV markers. However, their relative enrichment (profile) on EVs of specific cellular origins is not fully elucidated. We introduce LuminEV, a novel version of the Luminex assay for the multiplexed analysis of EV surface proteins. Optimized LuminEV reagents enable direct, specific, and sensitive measurements of EV markers in biofluids and in culture supernatants, bypassing EV isolation step. LuminEV assay for CD9, CD63, and CD81 was validated by comparing simplex and multiplex measurements, establishing linearity, spike-in recovery, inter- and intra-assay precision, and reproducibility between operators. LuminEV measurements of CD9, CD63, and CD81 in conditioned media from 15 cell lines revealed strong variations between cell types and showed high sensitivity, which enabled EV detection without prior concentration. Using tetraspanin levels as a readout, we noted suppression and induction of EV release from the cultured cells by GW6869 and monensin. Measurement of EV CD9, CD63, and CD81 in blood plasma from 70 disease-free donors showed respective abundance of 72, 16, and 12%. CD63 displayed weak, albeit significant, negative correlation with age and was slightly lower in female samples. The assay was then used to detect cell type-specific EV surface markers, including CD235a (erythrocytes), GAP43 (neurons), and CD68 (macrophages), and to detect differences in tetraspanin profiles between healthy and diseased donors. In summary, LuminEV offers robust and sensitive approach for multiplexed assessment of EV surface proteins, to facilitate the research into EV biology, biomarker, and therapeutic applications.</p>","PeriodicalId":15811,"journal":{"name":"Journal of Extracellular Vesicles","volume":"13 11","pages":""},"PeriodicalIF":15.5,"publicationDate":"2024-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/jev2.70007","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142579595","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Correction to MAP kinase kinase 1 (MEK1) within extracellular vesicles inhibits tumour growth by promoting anti-tumour immunity","authors":"","doi":"10.1002/jev2.70010","DOIUrl":"https://doi.org/10.1002/jev2.70010","url":null,"abstract":"<p>Searles, S. C., Chen, W.-S., Yee, J. D., Lee, P., Lee, C. K., Caron, C., Mousovich-Neto, F., Matei, I., Lyden, D., & Bui, J. D. (2024). MAP kinase kinase 1 (MEK1) within extracellular vesicles inhibits tumour growth by promoting anti-tumour immunity. <i>Journal of Extracellular Vesicles</i>, 13, e12515. https://doi.org/10.1002/jev2.12515</p><p>In the originally-published article, author Felippe Mousovich-Neto's name was incorrectly given as Felippe Neto. The online version of the article has been corrected.</p><p>We apologize for this error.</p>","PeriodicalId":15811,"journal":{"name":"Journal of Extracellular Vesicles","volume":"13 11","pages":""},"PeriodicalIF":15.5,"publicationDate":"2024-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/jev2.70010","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142579582","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}