151
|
Pike S, Peng CC, Neviani P, Berry JL, Xu L. CD63/81 Small Extracellular Vesicles in the Aqueous Humor are Retinoblastoma Associated. Invest Ophthalmol Vis Sci 2023; 64:5. [PMID: 37410475 PMCID: PMC10337798 DOI: 10.1167/iovs.64.10.5] [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: 02/13/2023] [Accepted: 06/11/2023] [Indexed: 07/07/2023] Open
Abstract
Purpose Although biopsy is contraindicated in retinoblastoma (RB), the aqueous humor (AH) is a robust liquid biopsy source of molecular tumor information, facilitating biomarker discovery. Small extracellular vesicles (sEVs), promising biomarker candidates across multiple cancers, were recently identified in RB AH, but relationships between sEVs and RB clinical features are unknown. Methods We analyzed sEVs in 37 AH samples from 18 RB eyes of varying International Intraocular Retinoblastoma Classification (IIRC) groups and explored clinical correlations. Ten samples were collected at diagnosis (DX) and 27 during treatment (Tx). Unprocessed AH underwent Single Particle-Interferometric Reflectance Imaging Sensor (SP-IRIS) analysis for fluorescent particle count and tetraspanin immunophenotyping; counts were subsequentially converted to percentages for analysis. Results Comparing DX and Tx samples, a higher percentage of CD63/81+ sEVs was found in DX AH (16.3 ± 11.6% vs. 5.49 ± 3.67% P = 0.0009), with a more homogenous mono-CD63+ sEV population seen in Tx AH (43.5 ± 14.7% vs. 28.8 ± 9.38%, P = 0.0073). Among DX samples, CD63/81+ sEVs were most abundant in group E eyes (n = 2) compared to group D (n = 6) by count (2.75 × 105 ± 3.40 × 105 vs. 5.95 × 103 ± 8.16 × 103, P = 0.0006), and to group A + B (n = 2) by count (2.75 × 105 ± 3.40 × 105 vs. 2.73 × 102 ± 2.59 × 102, P = 0.0096) and percentage (32.1 ± 7.98% vs. 7.79 ± 0.02%, P = 0.0187). Conclusions CD63/81+ sEVs enrich AH from RB eyes before treatment and those with more significant tumor burden, suggesting they are tumor-derived. Future research into their cargo may reveal mechanisms of cellular communication via sEVs in RB and novel biomarkers.
Collapse
Affiliation(s)
- Sarah Pike
- The Vision Center, Children's Hospital Los Angeles, Los Angeles, California, United States
- USC Roski Eye Institute, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
| | - Chen-Ching Peng
- The Vision Center, Children's Hospital Los Angeles, Los Angeles, California, United States
| | - Paolo Neviani
- Extracellular Vesicle Core, Children's Hospital Los Angeles, Los Angeles, California, United States
| | - Jesse L. Berry
- The Vision Center, Children's Hospital Los Angeles, Los Angeles, California, United States
- USC Roski Eye Institute, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, United States
- Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
| | - Liya Xu
- The Vision Center, Children's Hospital Los Angeles, Los Angeles, California, United States
- USC Roski Eye Institute, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, United States
| |
Collapse
|
152
|
Benayas B, Morales J, Egea C, Armisén P, Yáñez‐Mó M. Optimization of extracellular vesicle isolation and their separation from lipoproteins by size exclusion chromatography. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e100. [PMID: 38939075 PMCID: PMC11080862 DOI: 10.1002/jex2.100] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 06/29/2024]
Abstract
Interest in the use of extracellular vesicles (EVs) as biomarkers of disease is rapidly growing. However, one main unsolved issue in the EV field is finding a technique able to eliminate non-EV contaminants present in biofluid samples in a one-step isolation protocol. Due to the expansion and value of size exclusion chromatography (SEC) as one of the best EV isolation methods, we have tested several agarose resins with different agarose percentages, bead sizes and crosslinking features to optimize EV isolation. For this optimization of SEC, we first employed conditioned media from a melanoma cell culture, a simpler sample in comparison to biological fluids, but which also contains abundant contaminants such as soluble protein and lipoproteins (LPPs). The distinct agaroses and the combinations of resins with different agarose percentages in the same column were tested. Soluble protein, EVs and LPPs levels from the different eluted fractions were quantitated by immunodetection or absorbance measurements. Samples were also analysed by NTA and TEM to verify the yield and the LPP contamination. Different percentages of agarose resins (2%, 4% and 6%) yielded samples with increasing LPP contamination respectively, which was not improved in the columns that combined them. Crosslinking of the agarose did not affect EV isolation yield nor the LPP contamination. In contrast, reducing the bead size greatly improved EV purity. We thus selected 4% Rapid Run Fine agarose beads as the resin that more efficiently isolated EVs with almost no contamination of other particles. Using blood plasma samples, this resin also demonstrated an improved capacity in the isolation of EVs from LPPs in comparison to the agaroses most commonly used in the field and differential ultracentrifugation.
Collapse
Affiliation(s)
- Beatriz Benayas
- Agarose Bead Technologies (ABT)Torrejón de ArdozMadridSpain
- Dept Biología MolecularUniversidad Autónoma de MadridIUBMCentro de Biología Molecular Severo Ochoa, IIS‐IPMadridSpain
| | - Joaquín Morales
- Dept Biología MolecularUniversidad Autónoma de MadridIUBMCentro de Biología Molecular Severo Ochoa, IIS‐IPMadridSpain
| | - Carolina Egea
- Agarose Bead Technologies (ABT)Torrejón de ArdozMadridSpain
| | - Pilar Armisén
- Agarose Bead Technologies (ABT)Torrejón de ArdozMadridSpain
| | - María Yáñez‐Mó
- Dept Biología MolecularUniversidad Autónoma de MadridIUBMCentro de Biología Molecular Severo Ochoa, IIS‐IPMadridSpain
| |
Collapse
|
153
|
Saftics A, Abuelreich S, Romano E, Ghaeli I, Jiang N, Spanos M, Lennon KM, Singh G, Das S, Van Keuren‐Jensen K, Jovanovic‐Talisman T. Single Extracellular VEsicle Nanoscopy. J Extracell Vesicles 2023; 12:e12346. [PMID: 37422692 PMCID: PMC10329735 DOI: 10.1002/jev2.12346] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 05/26/2023] [Accepted: 06/23/2023] [Indexed: 07/10/2023] Open
Abstract
Extracellular vesicles (EVs) and their cargo constitute novel biomarkers. EV subpopulations have been defined not only by abundant tetraspanins (e.g., CD9, CD63 and CD81) but also by specific markers derived from their source cells. However, it remains a challenge to robustly isolate and characterize EV subpopulations. Here, we combined affinity isolation with super-resolution imaging to comprehensively assess EV subpopulations from human plasma. Our Single Extracellular VEsicle Nanoscopy (SEVEN) assay successfully quantified the number of affinity-isolated EVs, their size, shape, molecular tetraspanin content, and heterogeneity. The number of detected tetraspanin-enriched EVs positively correlated with sample dilution in a 64-fold range (for SEC-enriched plasma) and a 50-fold range (for crude plasma). Importantly, SEVEN robustly detected EVs from as little as ∼0.1 μL of crude plasma. We further characterized the size, shape and molecular tetraspanin content (with corresponding heterogeneities) for CD9-, CD63- and CD81-enriched EV subpopulations. Finally, we assessed EVs from the plasma of four pancreatic ductal adenocarcinoma patients with resectable disease. Compared to healthy plasma, CD9-enriched EVs from patients were smaller while IGF1R-enriched EVs from patients were larger, rounder and contained more tetraspanin molecules, suggestive of a unique pancreatic cancer-enriched EV subpopulation. This study provides the method validation and demonstrates that SEVEN could be advanced into a platform for characterizing both disease-associated and organ-associated EV subpopulations.
Collapse
Affiliation(s)
- Andras Saftics
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Sarah Abuelreich
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Eugenia Romano
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Ima Ghaeli
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Nan Jiang
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Michail Spanos
- Cardiology Division and Corrigan Minehan Heart CenterMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Kathleen M. Lennon
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Gagandeep Singh
- Department of SurgeryCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Saumya Das
- Cardiology Division and Corrigan Minehan Heart CenterMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | | | - Tijana Jovanovic‐Talisman
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| |
Collapse
|
154
|
Yu H, Lu Y, Lan F, Wang Y, Hu C, Mao L, Wu D, Li F, Song H. Engineering Outer Membrane Vesicles to Increase Extracellular Electron Transfer of Shewanella oneidensis. ACS Synth Biol 2023; 12:1645-1656. [PMID: 37140342 DOI: 10.1021/acssynbio.2c00636] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Outer membrane vesicles (OMVs) of Gram-negative bacteria play an essential role in cellular physiology. The underlying regulatory mechanism of OMV formation and its impact on extracellular electron transfer (EET) in the model exoelectrogenShewanella oneidensis MR-1 remain unclear and have not been reported. To explore the regulatory mechanism of OMV formation, we used the CRISPR-dCas9 gene repression technology to reduce the crosslink between the peptidoglycan (PG) layer and the outer membrane, thus promoting the OMV formation. We screened the target genes that were potentially beneficial to the outer membrane bulge, which were classified into two modules: PG integrity module (Module 1) and outer membrane component module (Module 2). We found that downregulation of the penicillin-binding protein-encoding gene pbpC for peptidoglycan integrity (Module 1) and the N-acetyl-d-mannosamine dehydrogenase-encoding gene wbpP involved in lipopolysaccharide synthesis (Module 2) exhibited the highest production of OMVs and enabled the highest output power density of 331.3 ± 1.2 and 363.8 ± 9.9 mW m-2, 6.33- and 6.96-fold higher than that of the wild-typeS. oneidensis MR-1 (52.3 ± 0.6 mW m-2), respectively. To elucidate the specific impacts of OMV formation on EET, OMVs were isolated and quantified for UV-visible spectroscopy and heme staining characterization. Our study showed that abundant outer membrane c-type cytochromes (c-Cyts) including MtrC and OmcA and periplasmic c-Cyts were exposed on the surface or inside of OMVs, which were the vital constituents responsible for EET. Meanwhile, we found that the overproduction of OMVs could facilitate biofilm formation and increase biofilm conductivity. To the best of our knowledge, this study is the first to explore the mechanism of OMV formation and its correlation with EET of S. oneidensis, which paves the way for further study of OMV-mediated EET.
Collapse
Affiliation(s)
- Huan Yu
- Frontier Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin 300072, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Yujun Lu
- Frontier Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin 300072, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Fei Lan
- Frontier Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin 300072, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Yuxuan Wang
- Frontier Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin 300072, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Chaoning Hu
- Frontier Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin 300072, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Lingfeng Mao
- Frontier Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin 300072, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Deguang Wu
- Department of Brewing Engineering, Moutai Institute, Luban Ave, Renhuai 564507, Guizhou, China
| | - Feng Li
- Frontier Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin 300072, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Hao Song
- Frontier Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin 300072, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| |
Collapse
|
155
|
Wu M, Yang Q, Yang C, Han J, Liu H, Qiao L, Duan H, Xing L, Liu Q, Dong L, Wang Q, Zuo L. Characteristics of plasma exosomes in drug-resistant tuberculosis patients. Tuberculosis (Edinb) 2023; 141:102359. [PMID: 37329682 DOI: 10.1016/j.tube.2023.102359] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Increasing prevalence of drug-resistant tuberculosis (DR-TB) poses a major challenge to the early detection and effective control of tuberculosis (TB). Exosomes carrying proteins and nucleic acid mediate intercellular communication between host and pathogen including Mycobacterium tuberculosis. However, molecular events of exosomes indicating the status and development of DR-TB remain unknown. This study determined the proteomics of exosome in DR-TB and explored the potential pathogenesis of DR-TB. METHODS Plasma samples were collected from 17 DR-TB patients and 33 non-drug-resistant tuberculosis (NDR-TB) patients using grouped case-control study design. After exosomes of plasma were isolated and confirmed by compositional and morphological measurement for exosomal characteristics, a label-free quantitative proteomics of exosomes was performed and differential protein components were determined via bioinformatics analysis. RESULTS Compared with the NDR-TB group, we identified 16 up-regulated proteins and 10 down-regulated proteins in the DR-TB group. The down-regulated proteins were mainly apolipoproteins and mainly enriched in cholesterol metabolism-related pathways. Apolipoproteins family including APOA1, APOB, APOC1 were key proteins in protein-protein interaction network. CONCLUSION Differentially expressed proteins in the exosomes may indicate the status of DR-TB from NDR-TB. Apolipoproteins family including APOA1, APOB, APOC1 may be involved in the pathogenesis of DR-TB by regulating cholesterol metabolism via exosomes.
Collapse
Affiliation(s)
- Mingrui Wu
- Key Laboratory of Cellular Physiology, Ministry of Education, The Department of Physiology, School of Basic Sciences, Shanxi Medical University, Taiyuan, 030001, China
| | - Qianwei Yang
- Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
| | - Caiting Yang
- Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
| | - Jie Han
- Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
| | - Hai Liu
- Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
| | - Lingran Qiao
- Key Laboratory of Cellular Physiology, Ministry of Education, The Department of Physiology, School of Basic Sciences, Shanxi Medical University, Taiyuan, 030001, China
| | - Huiping Duan
- The Fourth People's Hospital of Taiyuan, Taiyuan, 030024, China
| | - Li Xing
- Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
| | - Qunqun Liu
- The Fourth People's Hospital of Taiyuan, Taiyuan, 030024, China
| | - Li Dong
- Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China.
| | - Quanhong Wang
- The Fourth People's Hospital of Taiyuan, Taiyuan, 030024, China.
| | - Lin Zuo
- Key Laboratory of Cellular Physiology, Ministry of Education, The Department of Physiology, School of Basic Sciences, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
156
|
Hessvik NP, Sagini K, Romero S, Ramirez-Garrastacho M, Rodriguez M, Tutturen AEV, Kvalvaag A, Stang E, Brech A, Sandvig K, Llorente A. siRNA screening reveals that SNAP29 contributes to exosome release. Cell Mol Life Sci 2023; 80:177. [PMID: 37285022 PMCID: PMC10247572 DOI: 10.1007/s00018-023-04822-8] [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: 12/27/2022] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/08/2023]
Abstract
Cells release extracellular vesicles (EVs) of different sizes. Small EVs (< 200 nm) can originate from the fusion of multivesicular bodies with the plasma membrane, i.e. exosomes, and from budding of the plasma membrane, i.e. small ectosomes. To investigate the molecular machinery required for the release of small EVs, we developed a sensitive assay based on incorporation of radioactive cholesterol in EV membranes and used it in a siRNA screening. The screening showed that depletion of several SNARE proteins affected the release of small EVs. We focused on SNAP29, VAMP8, syntaxin 2, syntaxin 3 and syntaxin 18, the depletion of which reduced the release of small EVs. Importantly, this result was verified using gold standard techniques. SNAP29 depletion resulted in the largest effect and was further investigated. Immunoblotting analysis of small EVs showed that the release of several proteins considered to be associated with exosomes like syntenin, CD63 and Tsg101 was reduced, while the level of several proteins that have been shown to be released in ectosomes (annexins) or by secretory autophagy (LC3B and p62) was not affected by SNAP29 depletion. Moreover, these proteins appeared in different fractions when the EV samples were further separated by a density gradient. These results suggest that SNAP29 depletion mainly affects the secretion of exosomes. To investigate how SNAP29 affects exosome release, we used microscopy to study the distribution of MBVs using CD63 labelling and CD63-pHluorin to detect fusion events of MVBs with the plasma membrane. SNAP29 depletion caused a redistribution of CD63-labelled compartments but did not change the number of fusion events. Further experiments are therefore needed to fully understand the function of SNAP29. To conclude, we have developed a novel screening assay that has allowed us to identify several SNAREs involved in the release of small EVs.
Collapse
Affiliation(s)
- Nina Pettersen Hessvik
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Krizia Sagini
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Silvana Romero
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Manuel Ramirez-Garrastacho
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Marta Rodriguez
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Pathology Department, IIS-Fundación Jiménez Díaz-UAM, Center for the Biomedical Research Network in Oncology, CIBERONC, Madrid, Spain
| | | | - Audun Kvalvaag
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Espen Stang
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Andreas Brech
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway.
- Department for Mechanical, Electronics and Chemical Engineering, Oslo Metropolitan University, Oslo, Norway.
| |
Collapse
|
157
|
Paluschinski M, Loosen S, Kordes C, Keitel V, Kuebart A, Brandenburger T, Schöler D, Wammers M, Neumann UP, Luedde T, Castoldi M. Extracellular Vesicles as Markers of Liver Function: Optimized Workflow for Biomarker Identification in Liver Disease. Int J Mol Sci 2023; 24:9631. [PMID: 37298585 PMCID: PMC10253831 DOI: 10.3390/ijms24119631] [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: 05/09/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Liver diseases represent a significant global health burden, necessitating the development of reliable biomarkers for early detection, prognosis, and therapeutic monitoring. Extracellular vesicles (EVs) have emerged as promising candidates for liver disease biomarkers due to their unique cargo composition, stability, and accessibility in various biological fluids. In this study, we present an optimized workflow for the identification of EVs-based biomarkers in liver disease, encompassing EVs isolation, characterization, cargo analysis, and biomarker validation. Here we show that the levels of microRNAs miR-10a, miR-21, miR-142-3p, miR-150, and miR-223 were different among EVs isolated from patients with nonalcoholic fatty liver disease and autoimmune hepatitis. In addition, IL2, IL8, and interferon-gamma were found to be increased in EVs isolated from patients with cholangiocarcinoma compared with healthy controls. By implementing this optimized workflow, researchers and clinicians can improve the identification and utilization of EVs-based biomarkers, ultimately enhancing liver disease diagnosis, prognosis, and personalized treatment strategies.
Collapse
Affiliation(s)
- Martha Paluschinski
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany; (M.P.); (S.L.); (C.K.); (V.K.); (D.S.); (M.W.); (T.L.)
| | - Sven Loosen
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany; (M.P.); (S.L.); (C.K.); (V.K.); (D.S.); (M.W.); (T.L.)
| | - Claus Kordes
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany; (M.P.); (S.L.); (C.K.); (V.K.); (D.S.); (M.W.); (T.L.)
| | - Verena Keitel
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany; (M.P.); (S.L.); (C.K.); (V.K.); (D.S.); (M.W.); (T.L.)
| | - Anne Kuebart
- Department of Anesthesiology, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany; (A.K.); (T.B.)
| | - Timo Brandenburger
- Department of Anesthesiology, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany; (A.K.); (T.B.)
| | - David Schöler
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany; (M.P.); (S.L.); (C.K.); (V.K.); (D.S.); (M.W.); (T.L.)
| | - Marianne Wammers
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany; (M.P.); (S.L.); (C.K.); (V.K.); (D.S.); (M.W.); (T.L.)
| | - Ulf P. Neumann
- Visceral and Transplant Surgery, University Hospital RWTH Aachen, 52074 Aachen, Germany;
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany; (M.P.); (S.L.); (C.K.); (V.K.); (D.S.); (M.W.); (T.L.)
| | - Mirco Castoldi
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany; (M.P.); (S.L.); (C.K.); (V.K.); (D.S.); (M.W.); (T.L.)
| |
Collapse
|
158
|
Dhar R, Gorai S, Devi A, Muthusamy R, Alexiou A, Papadakis M. Decoding of exosome heterogeneity for cancer theranostics. Clin Transl Med 2023; 13:e1288. [PMID: 37292029 PMCID: PMC10251134 DOI: 10.1002/ctm2.1288] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 05/20/2023] [Accepted: 05/22/2023] [Indexed: 06/10/2023] Open
Affiliation(s)
- Rajib Dhar
- Department of Genetic EngineeringCancer and Stem Cell Biology LaboratorySRM Institute of Science and TechnologyKattankulathurIndia
| | - Sukhamoy Gorai
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Arikketh Devi
- Department of Genetic EngineeringCancer and Stem Cell Biology LaboratorySRM Institute of Science and TechnologyKattankulathurIndia
| | - Raman Muthusamy
- Department of MicrobiologyCentre for Infectious DiseasesSaveetha Dental CollegeChennaiIndia
| | - Athanasios Alexiou
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamAustralia
- AFNP MedWienAustria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten‐Herdecke, Heusnerstrasse 40University of Witten‐HerdeckeWuppertalGermany
| |
Collapse
|
159
|
Fusco P, Fietta A, Esposito MR, Zanella L, Micheli S, Bastianello A, Bova L, Borile G, Germano G, Cimetta E. miR-210-3p enriched extracellular vesicles from hypoxic neuroblastoma cells stimulate migration and invasion of target cells. Cell Biosci 2023; 13:89. [PMID: 37202777 DOI: 10.1186/s13578-023-01045-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/03/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Tumor hypoxia stimulates release of extracellular vesicles (EVs) that facilitate short- and long-range intercellular communication and metastatization. Albeit hypoxia and EVs release are known features of Neuroblastoma (NB), a metastasis-prone childhood malignancy of the sympathetic nervous system, whether hypoxic EVs can facilitate NB dissemination is unclear. METHODS Here we isolated and characterized EVs from normoxic and hypoxic NB cell culture supernatants and performed microRNA (miRNA) cargo analysis to identify key mediators of EVs biological effects. We then validated if EVs promote pro-metastatic features both in vitro and in an in vivo zebrafish model. RESULTS EVs from NB cells cultured at different oxygen tensions did not differ for type and abundance of surface markers nor for biophysical properties. However, EVs derived from hypoxic NB cells (hEVs) were more potent than their normoxic counterpart in inducing NB cells migration and colony formation. miR-210-3p was the most abundant miRNA in the cargo of hEVs; mechanistically, overexpression of miR-210-3p in normoxic EVs conferred them pro-metastatic features, whereas miR-210-3p silencing suppressed the metastatic ability of hypoxic EVs both in vitro and in vivo. CONCLUSION Our data identify a role for hypoxic EVs and their miR-210-3p cargo enrichment in the cellular and microenvironmental changes favoring NB dissemination.
Collapse
Affiliation(s)
- Pina Fusco
- Department of Industrial Engineering (DII), University of Padua, Via Marzolo 9, 35131, Padua, Italy
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy
| | - Anna Fietta
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy
| | - Maria Rosaria Esposito
- Department of Industrial Engineering (DII), University of Padua, Via Marzolo 9, 35131, Padua, Italy
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy
| | - Luca Zanella
- Department of Industrial Engineering (DII), University of Padua, Via Marzolo 9, 35131, Padua, Italy
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy
| | - Sara Micheli
- Department of Industrial Engineering (DII), University of Padua, Via Marzolo 9, 35131, Padua, Italy
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy
| | - Angelica Bastianello
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy
| | - Lorenzo Bova
- Department of Industrial Engineering (DII), University of Padua, Via Marzolo 9, 35131, Padua, Italy
| | - Giulia Borile
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy
| | - Giuseppe Germano
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy
| | - Elisa Cimetta
- Department of Industrial Engineering (DII), University of Padua, Via Marzolo 9, 35131, Padua, Italy.
- Fondazione Istituto Di Ricerca Pediatrica Città Della Speranza (IRP), Corso Stati Uniti 4, 35127, Padua, Italy.
| |
Collapse
|
160
|
Buchacher T, Digruber A, Kranzler M, Del Favero G, Ehling-Schulz M. Bacillus cereus extracellular vesicles act as shuttles for biologically active multicomponent enterotoxins. Cell Commun Signal 2023; 21:112. [PMID: 37189133 PMCID: PMC10184354 DOI: 10.1186/s12964-023-01132-1] [Citation(s) in RCA: 12] [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: 03/14/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) from Gram-positive bacteria have gained considerable importance as a novel transport system of virulence factors in host-pathogen interactions. Bacillus cereus is a Gram-positive human pathogen, causing gastrointestinal toxemia as well as local and systemic infections. The pathogenicity of enteropathogenic B. cereus has been linked to a collection of virulence factors and exotoxins. Nevertheless, the exact mechanism of virulence factor secretion and delivery to target cells is poorly understood. RESULTS Here, we investigate the production and characterization of enterotoxin-associated EVs from the enteropathogenic B. cereus strain NVH0075-95 by using a proteomics approach and studied their interaction with human host cells in vitro. For the first time, comprehensive analyses of B. cereus EV proteins revealed virulence-associated factors, such as sphingomyelinase, phospholipase C, and the three-component enterotoxin Nhe. The detection of Nhe subunits was confirmed by immunoblotting, showing that the low abundant subunit NheC was exclusively detected in EVs as compared to vesicle-free supernatant. Cholesterol-dependent fusion and predominantly dynamin-mediated endocytosis of B. cereus EVs with the plasma membrane of intestinal epithelial Caco2 cells represent entry routes for delivery of Nhe components to host cells, which was assessed by confocal microscopy and finally led to delayed cytotoxicity. Furthermore, we could show that B. cereus EVs elicit an inflammatory response in human monocytes and contribute to erythrocyte lysis via a cooperative interaction of enterotoxin Nhe and sphingomyelinase. CONCLUSION Our results provide insights into the interaction of EVs from B. cereus with human host cells and add a new layer of complexity to our understanding of multicomponent enterotoxin assembly, offering new opportunities to decipher molecular processes involved in disease development. Video Abstract.
Collapse
Affiliation(s)
- Tanja Buchacher
- Functional Microbiology, Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Astrid Digruber
- Functional Microbiology, Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Markus Kranzler
- Functional Microbiology, Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Monika Ehling-Schulz
- Functional Microbiology, Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria.
| |
Collapse
|
161
|
Steć A, Chodkowska M, Kasprzyk-Pochopień J, Mielczarek P, Piekoszewski W, Lewczuk B, Płoska A, Kalinowski L, Wielgomas B, Dziomba S. Isolation of Citrus lemon extracellular vesicles: Development and process control using capillary electrophoresis. Food Chem 2023; 424:136333. [PMID: 37201469 DOI: 10.1016/j.foodchem.2023.136333] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/20/2023]
Abstract
A new and scalable method for the isolation of extracellular vesicles (EV) from Citrus lemon juice samples was developed. The methodology included preliminary preconcentration of the sample using ultrafiltration (UF) followed by size-exclusion chromatography (SEC) purification and final preconcentration of the eluates. Transmission electron microscopy and proteomic analysis showed that isolates contained exosome-like vesicles, exocyst-positive organelle (EXPO), and microvesicles. The efficiency of certain isolation steps was evaluated with total protein content assay (bicinchoninic acid assay, BCA), nanoparticles tracking analysis (NTA), and capillary electrophoresis (CE). A good correlation between CE, BCA, and NTA results was shown. The application of CE enabled the detection of soluble contaminants, macromolecular aggregates, and vesicles' heterogeneity. The fluorescent staining of encapsulated nucleic acids was proposed for the identity confirmation of EV detected in CE. The study demonstrates the CE as a comprehensive tool for monitoring of the EV isolation process.
Collapse
Affiliation(s)
- Aleksandra Steć
- Department of Toxicology, Faculty of Pharmacy, Medical University of Gdansk, 107 Hallera Street, 80-416 Gdansk, Poland
| | - Martyna Chodkowska
- Department of Toxicology, Faculty of Pharmacy, Medical University of Gdansk, 107 Hallera Street, 80-416 Gdansk, Poland
| | - Joanna Kasprzyk-Pochopień
- Laboratory of High-Resolution Mass Spectrometry, Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Street, 30-387 Krakow, Poland
| | - Przemyslaw Mielczarek
- AGH University of Science and Technology, Faculty of Materials Science and Ceramics, Department of Analytical Chemistry and Biochemistry, 30 Mickiewicza Avenue, 30-059 Krakow, Poland
| | - Wojciech Piekoszewski
- Laboratory of High-Resolution Mass Spectrometry, Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Street, 30-387 Krakow, Poland; Department of Analytical Chemistry, Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Street, 30-387 Krakow, Poland
| | - Bogdan Lewczuk
- University of Warmia and Mazury in Olsztyn, Department of Histology and Embryology, 13 Oczapowskiego Street, 10-713 Olsztyn, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.pl, Faculty of Pharmacy, Medical University of Gdansk, 7 Debinki Street, 80-211 Gdansk, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.pl, Faculty of Pharmacy, Medical University of Gdansk, 7 Debinki Street, 80-211 Gdansk, Poland; BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 11/12 Narutowicza Street, 80-233 Gdansk, Poland
| | - Bartosz Wielgomas
- Department of Toxicology, Faculty of Pharmacy, Medical University of Gdansk, 107 Hallera Street, 80-416 Gdansk, Poland
| | - Szymon Dziomba
- Department of Toxicology, Faculty of Pharmacy, Medical University of Gdansk, 107 Hallera Street, 80-416 Gdansk, Poland.
| |
Collapse
|
162
|
Takeda M, Akamatsu S, Kita Y, Goto T, Kobayashi T. The Roles of Extracellular Vesicles in the Progression of Renal Cell Carcinoma and Their Potential for Future Clinical Application. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13101611. [PMID: 37242027 DOI: 10.3390/nano13101611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023]
Abstract
Renal cell carcinoma (RCC) is the most common type of kidney cancer and is thought to originate from renal tubular epithelial cells. Extracellular vesicles (EVs) are nanosized lipid bilayer vesicles that are secreted into extracellular spaces by nearly all cell types, including cancer cells and non-cancerous cells. EVs are involved in multiple steps of RCC progression, such as local invasion, host immune modulation, drug resistance, and metastasis. Therefore, EVs secreted from RCC are attracting rapidly increasing attention from researchers. In this review, we highlight the mechanism by which RCC-derived EVs lead to disease progression as well as the potential and challenges related to the clinical implications of EV-based diagnostics and therapeutics.
Collapse
Affiliation(s)
- Masashi Takeda
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Shusuke Akamatsu
- Department of Urology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Yuki Kita
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Takayuki Goto
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Takashi Kobayashi
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
163
|
Blanco NG, Machado NM, Castro LL, Antunes MA, Takiya CM, Trugilho MRO, Silva LR, Paes Leme AF, Domingues RR, Pauletti BA, Miranda BT, Silva JD, Dos Santos CC, Silva PL, Rocco PRM, Cruz FF. Extracellular Vesicles from Different Sources of Mesenchymal Stromal Cells Have Distinct Effects on Lung and Distal Organs in Experimental Sepsis. Int J Mol Sci 2023; 24:ijms24098234. [PMID: 37175936 PMCID: PMC10179270 DOI: 10.3390/ijms24098234] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 04/24/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
The effects of the administration of mesenchymal stromal cells (MSC) may vary according to the source. We hypothesized that MSC-derived extracellular vesicles (EVs) obtained from bone marrow (BM), adipose (AD), or lung (L) tissues may also lead to different effects in sepsis. We profiled the proteome from EVs as a first step toward understanding their mechanisms of action. Polymicrobial sepsis was induced in C57BL/6 mice by cecal ligation and puncture (SEPSIS) and SHAM (control) animals only underwent laparotomy. Twenty-four hours after surgery, animals in the SEPSIS group were randomized to receive saline or 3 × 106 MSC-derived EVs from BM, AD, or L. The diffuse alveolar damage was decreased with EVs from all three sources. In kidneys, BM-, AD-, and L-EVs reduced edema and expression of interleukin-18. Kidney injury molecule-1 expression decreased only in BM- and L-EVs groups. In the liver, only BM-EVs reduced congestion and cell infiltration. The size and number of EVs from different sources were not different, but the proteome of the EVs differed. BM-EVs were enriched for anti-inflammatory proteins compared with AD-EVs and L-EVs. In conclusion, BM-EVs were associated with less organ damage compared with the other sources of EVs, which may be related to differences detected in their proteome.
Collapse
Affiliation(s)
- Natália G Blanco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro 21941-902, RJ, Brazil
| | - Natália M Machado
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro 21941-902, RJ, Brazil
| | - Ligia L Castro
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro 21941-902, RJ, Brazil
| | - Mariana A Antunes
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro 21941-902, RJ, Brazil
| | - Christina M Takiya
- Laboratory of Immunopathology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Monique R O Trugilho
- Toxinology Laboratory, Center for Technological Development Health, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil
| | - Luana R Silva
- Toxinology Laboratory, Center for Technological Development Health, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil
| | - Adriana F Paes Leme
- Mass Spectrometry Laboratory, Brazilian Bioscience National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials, Campinas 13083-970, SP, Brazil
| | - Romênia R Domingues
- Mass Spectrometry Laboratory, Brazilian Bioscience National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials, Campinas 13083-970, SP, Brazil
| | - Bianca A Pauletti
- Mass Spectrometry Laboratory, Brazilian Bioscience National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials, Campinas 13083-970, SP, Brazil
| | - Beatriz T Miranda
- Laboratory of Cellular and Molecular Cardiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Johnatas D Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro 21941-902, RJ, Brazil
| | - Claudia C Dos Santos
- The Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, 209 Victoria Street, Toronto, ON M5B 1T8, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Pedro L Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro 21941-902, RJ, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro 21941-902, RJ, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro 21941-902, RJ, Brazil
| |
Collapse
|
164
|
Wang L, Bank MS, Rinklebe J, Hou D. Plastic-Rock Complexes as Hotspots for Microplastic Generation. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:7009-7017. [PMID: 37010423 DOI: 10.1021/acs.est.3c00662] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Discarded plastics and microplastics (MPs) in the environment are considered emerging contaminants and indicators of the Anthropocene epoch. This study reports the discovery of a new type of plastic material in the environment─plastic-rock complexes─formed when plastic debris irreversibly sorbs onto the parent rock after historical flooding events. These complexes consist of low-density polyethylene (LDPE) or polypropylene (PP) films stuck onto quartz-dominated mineral matrices. These plastic-rock complexes serve as hotspots for MP generation, as evidenced by laboratory wet-dry cycling tests. Over 1.03 × 108 and 1.28 × 108 items·m-2 MPs were generated in a zero-order mode from the LDPE- and PP-rock complexes, respectively, following 10 wet-dry cycles. The speed of MP generation was 4-5 orders of magnitude higher than that in landfills, 2-3 orders of magnitude higher than that in seawater, and >1 order of magnitude higher than that in marine sediment as compared with previously reported data. Results from this investigation provide strong direct evidence of anthropogenic waste entering geological cycles and inducing potential ecological risks that may be exacerbated by climate change conditions such as flooding events. Future research should evaluate this phenomenon regarding ecosystem fluxes, fate, and transport and impacts of plastic pollution.
Collapse
Affiliation(s)
- Liuwei Wang
- School of Environment, Tsinghua University, Beijing 100084, China
| | - Michael S Bank
- Institute of Marine Research, Bergen NO-5817, Norway
- University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Jörg Rinklebe
- Laboratory of Soil- and Groundwater-Management, Institute of Foundation Engineering, Water- and Waste-Management, School of Architecture and Civil Engineering, University of Wuppertal, Pauluskirchstraße 7, Wuppertal 42285, Germany
| | - Deyi Hou
- School of Environment, Tsinghua University, Beijing 100084, China
| |
Collapse
|
165
|
Zhang Q, Jeppesen DK, Higginbotham JN, Franklin JL, Coffey RJ. Comprehensive isolation of extracellular vesicles and nanoparticles. Nat Protoc 2023; 18:1462-1487. [PMID: 36914899 PMCID: PMC10445291 DOI: 10.1038/s41596-023-00811-0] [Citation(s) in RCA: 114] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 01/10/2023] [Indexed: 03/16/2023]
Abstract
There is an increasing appreciation for the heterogeneous nature of extracellular vesicles (EVs). In addition, two nonvesicular extracellular nanoparticles (NVEPs), exomeres and supermeres, have been discovered recently that are enriched in many cargo previously ascribed to EVs. The EV field has largely focused on EV isolation and characterization, while studies on NVEPs are limited. At this juncture, it is critically important to have robust and reliable methods to separate distinct populations of EVs and NVEPs to assign cargo to their correct carrier. Here, we provide a comprehensive step-by-step protocol for sequential isolation of large and small EVs, nonvesicular fractions, exomeres and supermeres from the same starting material. We describe in detail the use of differential ultracentrifugation, filtration, concentration and high-resolution density-gradient fractionation to obtain purified fractions of distinct populations of EVs and NVEPs. This protocol allows assignment and enrichment of a biomolecule of interest to its specific extracellular compartment. Compared to other isolation methods, our protocol has unique advantages, including high purity and reproducibility, with minimal expertise required. The protocol can be applied to purification of EVs and NVEPs from cell culture medium and human plasma and requires ~72 h to complete. Adoption of this protocol will help translational investigators identify potential circulating biomarkers and therapeutic targets for a host of human diseases and allow basic scientists to better understand EV and NVEP biogenesis and function. Overall, this protocol will allow those interested in isolating EVs and extracellular particles to advance scientific inquiry to answer outstanding questions in the field.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dennis K Jeppesen
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James N Higginbotham
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeffrey L Franklin
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Robert J Coffey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
166
|
Diehl JN, Ray A, Collins LB, Peterson A, Alexander KC, Boutros JG, Ikonomidis JS, Akerman AW. A standardized method for plasma extracellular vesicle isolation and size distribution analysis. PLoS One 2023; 18:e0284875. [PMID: 37115777 PMCID: PMC10146456 DOI: 10.1371/journal.pone.0284875] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
The following protocol describes our workflow for isolation and quantification of plasma extracellular vesicles (EVs). It requires limited sample volume so that the scientific value of specimens is maximized. These steps include isolation of vesicles by automated size exclusion chromatography and quantification by tunable resistive pulse sensing. This workflow optimizes reproducibility by minimizing variations in processing, handling, and storage of EVs. EVs have significant diagnostic and therapeutic potential, but clinical application is limited by disparate methods of data collection. This standardized protocol is scalable and ensures efficient recovery of physiologically intact EVs that may be used in a variety of downstream biochemical and functional analyses. Simultaneous measurement quantifies EV concentration and size distribution absolutely. Absolute quantification corrects for variations in EV number and size, offering a novel method of standardization in downstream applications.
Collapse
Affiliation(s)
- J. Nathaniel Diehl
- University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Amelia Ray
- University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Lauren B. Collins
- University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Andrew Peterson
- Department of Surgery, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina, United States of America
- Division of Cardiothoracic Surgery, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Kyle C. Alexander
- Department of Surgery, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina, United States of America
- Division of Cardiothoracic Surgery, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jacob G. Boutros
- Campbell University School of Osteopathic Medicine, Lillington, North Carolina, United States of America
| | - John S. Ikonomidis
- Department of Surgery, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina, United States of America
- Division of Cardiothoracic Surgery, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Adam W. Akerman
- Department of Surgery, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina, United States of America
- Division of Cardiothoracic Surgery, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
167
|
Huber MJ, Ivleva NP, Booth AM, Beer I, Bianchi I, Drexel R, Geiss O, Mehn D, Meier F, Molska A, Parot J, Sørensen L, Vella G, Prina-Mello A, Vogel R, Caputo F. Physicochemical characterization and quantification of nanoplastics: applicability, limitations and complementarity of batch and fractionation methods. Anal Bioanal Chem 2023:10.1007/s00216-023-04689-5. [PMID: 37106123 DOI: 10.1007/s00216-023-04689-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023]
Abstract
A comprehensive physicochemical characterization of heterogeneous nanoplastic (NPL) samples remains an analytical challenge requiring a combination of orthogonal measurement techniques to improve the accuracy and robustness of the results. Here, batch methods, including dynamic light scattering (DLS), nanoparticle tracking analysis (NTA), tunable resistive pulse sensing (TRPS), transmission electron microscopy (TEM), and scanning electron microscopy (SEM), as well as separation/fractionation methods such as centrifugal liquid sedimentation (CLS) and field-flow fractionation (FFF)-multi-angle light scattering (MALS) combined with pyrolysis gas chromatography mass spectrometry (pyGC-MS) or Raman microspectroscopy (RM) were evaluated for NPL size, shape, and chemical composition measurements and for quantification. A set of representative/test particles of different chemical natures, including (i) polydisperse polyethylene (PE), (ii) (doped) polystyrene (PS) NPLs, (iii) titanium dioxide, and (iv) iron oxide nanoparticles (spherical and elongated), was used to assess the applicability and limitations of the selected methodologies. Particle sizes and number-based concentrations obtained by orthogonal batch methods (DLS, NTA, TRPS) were comparable for monodisperse spherical samples, while higher deviations were observed for polydisperse, agglomerated samples and for non-spherical particles, especially for light scattering methods. CLS and TRPS offer further insight with increased size resolution, while detailed morphological information can be derived by electron microscopy (EM)-based approaches. Combined techniques such as FFF coupled to MALS and RM can provide complementary information on physical and chemical properties by online measurements, while pyGC-MS analysis of FFF fractions can be used for the identification of polymer particles (vs. inorganic particles) and for their offline (semi)quantification. However, NPL analysis in complex samples will continue to present a serious challenge for the evaluated techniques without significant improvements in sample preparation.
Collapse
Affiliation(s)
- Maximilian J Huber
- Institute of Water Chemistry (IWC), Chair of Analytical Chemistry and Water Chemistry, School of Natural Sciences (NAT, Dep. Chemistry), Technical University of Munich (TUM), Lichtenbergstr. 4, 85748, Garching, Germany
| | - Natalia P Ivleva
- Institute of Water Chemistry (IWC), Chair of Analytical Chemistry and Water Chemistry, School of Natural Sciences (NAT, Dep. Chemistry), Technical University of Munich (TUM), Lichtenbergstr. 4, 85748, Garching, Germany.
| | - Andy M Booth
- Department of Climate and Environment, SINTEF Ocean AS, Trondheim, Norway.
| | - Irina Beer
- Institute of Water Chemistry (IWC), Chair of Analytical Chemistry and Water Chemistry, School of Natural Sciences (NAT, Dep. Chemistry), Technical University of Munich (TUM), Lichtenbergstr. 4, 85748, Garching, Germany
| | - Ivana Bianchi
- Joint Research Centre (JRC), European Commission, Ispra, Italy
| | | | - Otmar Geiss
- Joint Research Centre (JRC), European Commission, Ispra, Italy
| | - Dora Mehn
- Joint Research Centre (JRC), European Commission, Ispra, Italy
| | | | - Alicja Molska
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Jeremie Parot
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Lisbet Sørensen
- Department of Climate and Environment, SINTEF Ocean AS, Trondheim, Norway
| | - Gabriele Vella
- Laboratory of Biological Characterization for Advanced Materials (LBCAM), Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Adriele Prina-Mello
- Laboratory of Biological Characterization for Advanced Materials (LBCAM), Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Robert Vogel
- School of Mathematics and Physics, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Fanny Caputo
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway.
- Laboratoire National de Métrologie et d'Essais, Paris, France.
| |
Collapse
|
168
|
Almeida C, Teixeira AL, Dias F, Morais M, Medeiros R. Extracellular Vesicles as Potential Therapeutic Messengers in Cancer Management. BIOLOGY 2023; 12:biology12050665. [PMID: 37237479 DOI: 10.3390/biology12050665] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023]
Abstract
A deeper understanding of the communication mechanisms of tumor cells in a tumor microenvironment can improve the development of new therapeutic solutions, leading to a more personalized approach. Recently, the field of extracellular vesicles (EVs) has drawn attention due to their key role in intercellular communication. EVs are nano-sized lipid bilayer vesicles that are secreted by all types of cells and can function as intermediators of intercellular communication with the ability to transfer different cargo (proteins, nucleic acids, sugar…) types among cells. This role of EVs is essential in a cancer context as it can affect tumor promotion and progression and contribute to the pre-metastatic niche establishment. Therefore, scientists from basic, translational, and clinical research areas are currently researching EVs with great expectations due to their potential to be used as clinical biomarkers, which are useful for disease diagnosis, prognosis, patient follow-up, or even as vehicles for drug delivery due to their natural carrier nature. The application of EVs presents numerous advantages as drug delivery vehicles, namely their capacity to overcome natural barriers, their inherent cell-targeting properties, and their stability in the circulation. In this review, we highlight the distinctive features of EVs, their application as efficient drug delivery systems, and their clinical applications.
Collapse
Affiliation(s)
- Cristina Almeida
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Rua Dr António Bernardino de Almeida, 4200-072 Porto, Portugal
- Research Department of the Portuguese League Against Cancer Regional Nucleus of the North (LPCC-NRNorte), Estrada da Circunvalação 6657, 4200-177 Porto, Portugal
| | - Ana Luísa Teixeira
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Rua Dr António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Francisca Dias
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Rua Dr António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Mariana Morais
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Rua Dr António Bernardino de Almeida, 4200-072 Porto, Portugal
- ICBAS School of Medicine and Biomedical Sciences, University of Porto (UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Rua Dr António Bernardino de Almeida, 4200-072 Porto, Portugal
- Research Department of the Portuguese League Against Cancer Regional Nucleus of the North (LPCC-NRNorte), Estrada da Circunvalação 6657, 4200-177 Porto, Portugal
- ICBAS School of Medicine and Biomedical Sciences, University of Porto (UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
- Fernando Pessoa Research, Innovation and Development Institute (I3ID FFP), Fernando Pessoa University (UFP), Praça 9 de Abril 349, 4249-004 Porto, Portugal
- Faculty of Medicine, University of Porto (FMUP), Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
169
|
S S, Camardo A, Dahal S, Ramamurthi A. Surface-Functionalized Stem Cell-Derived Extracellular Vesicles for Vascular Elastic Matrix Regenerative Repair. Mol Pharm 2023. [PMID: 37093652 DOI: 10.1021/acs.molpharmaceut.2c00769] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Extracellular vesicles (EVs) are nanosized vesicles that carry cell-specific biomolecular information. Our previous studies showed that adult human bone marrow mesenchymal stem cell (BM-MSC)-derived EVs provide antiproteolytic and proregenerative effects in cultures of smooth muscle cells (SMCs) derived from an elastase-infused rat abdominal aortic aneurysm (AAA) model, and this is promising toward their use as a therapeutic platform for naturally irreversible elastic matrix aberrations in the aortic wall. Since systemically administered EVs poorly home into sites of tissue injury, disease strategies to improve their affinity toward target tissues are of great significance for EV-based treatment strategies. Toward this goal, in this work, we developed a postisolation surface modification strategy to target MSC-derived EVs to the AAA wall. The EVs were surface-conjugated with a short, synthetic, azide-modified peptide sequence for targeted binding to cathepsin K (CatK), a cysteine protease overexpressed in the AAA wall. Conjugation was performed using a copper-free click chemistry method. We determined that such conjugation improved EV uptake into cultured aneurysmal SMCs in culture and their binding to the wall of matrix injured vessels ex vivo. The proregenerative and antiproteolytic effects of MSC-EVs on cultured rat aneurysmal SMCs were also unaffected following peptide conjugation. From this study, it appears that modification with short synthetic peptide sequences seems to be an effective strategy for improving the cell-specific uptake of EVs and may be effective in facilitating AAA-targeted therapy.
Collapse
Affiliation(s)
- Sajeesh S
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015-3027, United States
| | - Andrew Camardo
- Cleveland Clinic Foundation, 9620 Carnegie Ave. Cleveland, Ohio 44106, United States
| | - Shataakshi Dahal
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015-3027, United States
| | - Anand Ramamurthi
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015-3027, United States
| |
Collapse
|
170
|
Palacio PL, Pleet ML, Reátegui E, Magaña SM. Emerging role of extracellular vesicles in multiple sclerosis: From cellular surrogates to pathogenic mediators and beyond. J Neuroimmunol 2023; 377:578064. [PMID: 36934525 PMCID: PMC10124134 DOI: 10.1016/j.jneuroim.2023.578064] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/06/2023] [Accepted: 03/05/2023] [Indexed: 03/16/2023]
Abstract
Multiple Sclerosis (MS) is a chronic, inflammatory demyelinating disease of the central nervous system (CNS) driven by a complex interplay of genetic and environmental factors. While the therapeutic arsenal has expanded significantly for management of relapsing forms of MS, treatment of individuals with progressive MS is suboptimal. This treatment inequality is in part due to an incomplete understanding of pathomechanisms at different stages of the disease-underscoring the critical need for new biomarkers. Extracellular vesicles (EVs) and their bioactive cargo have emerged as endogenous nanoparticles with great theranostic potential-as diagnostic and prognostic biomarkers and ultimately as therapeutic candidates for precision nanotherapeutics. The goals of this review are to: 1) summarize the current data investigating the role of EVs and their bioactive cargo in MS pathogenesis, 2) provide a high level overview of advances and challenges in EV isolation and characterization for translational studies, and 3) conclude with future perspectives on this evolving field.
Collapse
Affiliation(s)
- Paola Loreto Palacio
- Department of Pediatrics, Division of Neurology, Nationwide Children's Hospital, Columbus, OH, USA
| | - Michelle L Pleet
- Viral Immunology Section, Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Eduardo Reátegui
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Setty M Magaña
- Department of Pediatrics, Division of Neurology, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
171
|
Abdelaziz MH, El Sawy EN, Abdelnaser A. A Novel Electrochemical Differentiation between Exosomal-RNA of Breast Cancer MCF7 and MCF7/ADR-Resistant Cells. Pharmaceuticals (Basel) 2023; 16:ph16040540. [PMID: 37111297 PMCID: PMC10145523 DOI: 10.3390/ph16040540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/09/2023] [Accepted: 03/23/2023] [Indexed: 04/29/2023] Open
Abstract
Cancer is considered one of the most burdensome diseases affecting lives and, hence, the economy. Breast cancer is one of the most common types of cancer. Patients with breast cancer are divided into two groups: one group responds to the chemotherapy, and the other group resists the chemotherapy. Unfortunately, the group which resists the chemotherapy is still suffering the pain associated with the severe side effects of the chemotherapy. Therefore, there is a critical need for a method to differentiate between both groups before the administration of the chemotherapy. Exosomes, the recently discovered nano-vesicles, are often used as cancer diagnostic biomarkers as their unique composition allows them to represent their parental cells, which makes them promising indicators for tumor prognosis. Exosomes contain proteins, lipids, and RNA that exist in most body fluids and are expelled by multiple cell types, including cancer cells. Furthermore, exosomal RNA has been significantly used as a promising biomarker for tumor prognosis. Herein, we have developed an electrochemical system that could successfully differentiate between MCF7 and MCF7/ADR depending on the exosomal RNA. The high sensitivity of the proposed electrochemical assay opens the door for further investigation that will address the other type of cancer cells.
Collapse
Affiliation(s)
- Mohammed H Abdelaziz
- Chemistry Department, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt
- Institute of Global Health and Human Ecology, The American University in Cairo, New Cairo 11835, Egypt
| | - Ehab N El Sawy
- Chemistry Department, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt
| | - Anwar Abdelnaser
- Institute of Global Health and Human Ecology, The American University in Cairo, New Cairo 11835, Egypt
| |
Collapse
|
172
|
Yi Q, Xu Z, Thakur A, Zhang K, Liang Q, Liu Y, Yan Y. Current understanding of plant-derived exosome-like nanoparticles in regulating the inflammatory response and immune system microenvironment. Pharmacol Res 2023; 190:106733. [PMID: 36931541 DOI: 10.1016/j.phrs.2023.106733] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
Natural compounds are widely used to prevent and treat various diseases due to their antioxidant and anti-inflammatory effects. As a kind of promising natural compound, plant-derived exosome-like nanoparticles (PELNs) are extracted from multivesicular bodies of various edible plants, including vegetables, foods, and fruits, and mainly regulate the cellular immune response to pathogen attacks. Moreover, PELNs could remarkably interfere with the dynamic imbalance between pro-inflammatory and anti-inflammatory effects, facilitating to maintain the homeostasis of cellular immune microenvironment. PELNs may serve as a better alternative to animal-derived exosomes (ADEs) owing to their widespread sources, cost-effectiveness, and easy accessibility. PELNs can mediate interspecies communication by transferring various cargoes such as proteins, lipids, and nucleic acids from plant cells to mammalian cells. This review summarizes the biogenesis, composition, and classification of exosomes; the common separation, purification, and characterization methods of PELNs, the potential advantages of PELNs over ADEs; and the anti-inflammatory and immunomodulatory functions of PELNs in various diseases including colitis, cancer, and inflammation-associated metabolic diseases. Additionally, the future perspectives of PELNs and the challenges associated with their clinical application are discussed.
Collapse
Affiliation(s)
- Qiaoli Yi
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Abhimanyu Thakur
- Pritzker School of Molecular Engineering, Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | - Kui Zhang
- State Key Laboratory of Silkworm Genome Biology, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Qiuju Liang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yuanhong Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| |
Collapse
|
173
|
Li T, Zhang L, Lu T, Zhu T, Feng C, Gao N, Liu F, Yu J, Chen K, Zhong J, Tang Q, Zhang Q, Deng X, Ren J, Zeng J, Zhou H, Zhu J. Engineered Extracellular Vesicle-Delivered CRISPR/CasRx as a Novel RNA Editing Tool. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206517. [PMID: 36727818 PMCID: PMC10074121 DOI: 10.1002/advs.202206517] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/13/2023] [Indexed: 06/10/2023]
Abstract
Engineered extracellular vesicles (EVs) are considered excellent delivery vehicles for a variety of therapeutic agents, including nucleic acids, proteins, drugs, and nanomaterials. Recently, several studies have indicated that clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated 9 (Cas9) delivered by EVs enable efficient DNA editing. However, an RNA editing tool delivered by EVs is still unavailable. Here, a signal peptide-optimized and EVs-delivered guide RNA (gRNA) and CRISPR/CasRx (Cas13d) system capable of rapidly inhibiting the expression of targeted genes with quick catabolism after performing their functions is developed. EVs with CRISPR/CasRx and tandem gRNAs targeting pivotal cytokines are further packed whose levels increase substantially over the course of acute inflammatory diseases and find that these engineered EVs inhibit macrophage activation in vitro. More importantly, this system attenuates lipopolysaccharide (LPS)-triggered acute lung injury and sepsis in the acute phase, mitigating organ damage and improving the prognosis in vivo. In summary, a potent tool is provided for short-acting RNA editing, which could be a powerful therapeutic platform for the treatment of acute diseases.
Collapse
Affiliation(s)
- Tianwen Li
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityNational Center for Neurological DisordersNational Key Laboratory for Medical NeurobiologyInstitutes of Brain ScienceShanghai Key Laboratory of Brain Function and RegenerationInstitute of NeurosurgeryMOE Frontiers Center for Brain ScienceShanghai200040China
| | - Liansheng Zhang
- Institute of NeuroscienceState Key Laboratory of NeuroscienceKey Laboratory of Primate NeurobiologyCAS Center for Excellence in Brain Science and Intelligence TechnologyShanghai Research Center for Brain Science and Brain‐Inspired IntelligenceShanghai Institutes for Biological SciencesChinese Academy of SciencesShanghai200031China
| | - Tao Lu
- Institute of NeuroscienceState Key Laboratory of NeuroscienceKey Laboratory of Primate NeurobiologyCAS Center for Excellence in Brain Science and Intelligence TechnologyShanghai Research Center for Brain Science and Brain‐Inspired IntelligenceShanghai Institutes for Biological SciencesChinese Academy of SciencesShanghai200031China
| | - Tongming Zhu
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityNational Center for Neurological DisordersNational Key Laboratory for Medical NeurobiologyInstitutes of Brain ScienceShanghai Key Laboratory of Brain Function and RegenerationInstitute of NeurosurgeryMOE Frontiers Center for Brain ScienceShanghai200040China
| | - Canbin Feng
- Institute of NeuroscienceState Key Laboratory of NeuroscienceKey Laboratory of Primate NeurobiologyCAS Center for Excellence in Brain Science and Intelligence TechnologyShanghai Research Center for Brain Science and Brain‐Inspired IntelligenceShanghai Institutes for Biological SciencesChinese Academy of SciencesShanghai200031China
| | - Ni Gao
- Institute of NeuroscienceState Key Laboratory of NeuroscienceKey Laboratory of Primate NeurobiologyCAS Center for Excellence in Brain Science and Intelligence TechnologyShanghai Research Center for Brain Science and Brain‐Inspired IntelligenceShanghai Institutes for Biological SciencesChinese Academy of SciencesShanghai200031China
| | - Fei Liu
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory DiseaseMolecular Diagnosis CenterDepartment of Pulmonary and Critical Care MedicineFirst Affiliated HospitalBengbu Medical CollegeNo. 287 Changhuai RoadBengbuAnhui233004China
| | - Jingyu Yu
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityNational Center for Neurological DisordersNational Key Laboratory for Medical NeurobiologyInstitutes of Brain ScienceShanghai Key Laboratory of Brain Function and RegenerationInstitute of NeurosurgeryMOE Frontiers Center for Brain ScienceShanghai200040China
| | - Kezhu Chen
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityNational Center for Neurological DisordersNational Key Laboratory for Medical NeurobiologyInstitutes of Brain ScienceShanghai Key Laboratory of Brain Function and RegenerationInstitute of NeurosurgeryMOE Frontiers Center for Brain ScienceShanghai200040China
| | - Junjie Zhong
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityNational Center for Neurological DisordersNational Key Laboratory for Medical NeurobiologyInstitutes of Brain ScienceShanghai Key Laboratory of Brain Function and RegenerationInstitute of NeurosurgeryMOE Frontiers Center for Brain ScienceShanghai200040China
| | - Qisheng Tang
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityNational Center for Neurological DisordersNational Key Laboratory for Medical NeurobiologyInstitutes of Brain ScienceShanghai Key Laboratory of Brain Function and RegenerationInstitute of NeurosurgeryMOE Frontiers Center for Brain ScienceShanghai200040China
| | - Quan Zhang
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityNational Center for Neurological DisordersNational Key Laboratory for Medical NeurobiologyInstitutes of Brain ScienceShanghai Key Laboratory of Brain Function and RegenerationInstitute of NeurosurgeryMOE Frontiers Center for Brain ScienceShanghai200040China
| | - Xiangyang Deng
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityNational Center for Neurological DisordersNational Key Laboratory for Medical NeurobiologyInstitutes of Brain ScienceShanghai Key Laboratory of Brain Function and RegenerationInstitute of NeurosurgeryMOE Frontiers Center for Brain ScienceShanghai200040China
| | - Junwei Ren
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityNational Center for Neurological DisordersNational Key Laboratory for Medical NeurobiologyInstitutes of Brain ScienceShanghai Key Laboratory of Brain Function and RegenerationInstitute of NeurosurgeryMOE Frontiers Center for Brain ScienceShanghai200040China
| | - Jun Zeng
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityNational Center for Neurological DisordersNational Key Laboratory for Medical NeurobiologyInstitutes of Brain ScienceShanghai Key Laboratory of Brain Function and RegenerationInstitute of NeurosurgeryMOE Frontiers Center for Brain ScienceShanghai200040China
| | - Haibo Zhou
- Institute of NeuroscienceState Key Laboratory of NeuroscienceKey Laboratory of Primate NeurobiologyCAS Center for Excellence in Brain Science and Intelligence TechnologyShanghai Research Center for Brain Science and Brain‐Inspired IntelligenceShanghai Institutes for Biological SciencesChinese Academy of SciencesShanghai200031China
| | - Jianhong Zhu
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityNational Center for Neurological DisordersNational Key Laboratory for Medical NeurobiologyInstitutes of Brain ScienceShanghai Key Laboratory of Brain Function and RegenerationInstitute of NeurosurgeryMOE Frontiers Center for Brain ScienceShanghai200040China
| |
Collapse
|
174
|
Smaldone G, Rosa E, Gallo E, Diaferia C, Morelli G, Stornaiuolo M, Accardo A. Caveolin-Mediated Internalization of Fmoc-FF Nanogels in Breast Cancer Cell Lines. Pharmaceutics 2023; 15:pharmaceutics15031026. [PMID: 36986886 PMCID: PMC10051563 DOI: 10.3390/pharmaceutics15031026] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
INTRODUCTION Hydrogel nanoparticles, also known as nanogels (NGs), have been recently proposed as alternative supramolecular vehicles for the delivery of biologically relevant molecules like anticancer drugs and contrast agents. The inner compartment of peptide based NGs can be opportunely modified according to the chemical features of the cargo, thus improving its loading and release. A full understanding of the intracellular mechanism involved in nanogel uptake by cancer cells and tissues would further contribute to the potential diagnostic and clinical applications of these nanocarriers, allowing the fine tuning of their selectivity, potency, and activity. The structural characterization of nanogels were assessed by Dynamic Light Scattering (DLS) and Nanoparticles Tracking Analysis (NTA) analysis. Cells viability of Fmoc-FF nanogels was evaluated by MTT assay on six breast cancer cell lines at different incubation times (24, 48, and 72 h) and peptide concentrations (in the range 6.25 × 10-4 ÷ 5·10-3 × wt%). The cell cycle and mechanisms involved in Fmoc-FF nanogels intracellular uptake were evaluated using flow cytometry and confocal analysis, respectively. Fmoc-FF nanogels, endowed with a diameter of ~130 nm and a zeta potential of ~-20.0/-25.0 mV, enter cancer cells via caveolae, mostly those responsible for albumin uptake. The specificity of the machinery used by Fmoc-FF nanogels confers a selectivity toward cancer cell lines overexpressing the protein caveolin1 and efficiently performing caveolae-mediated endocytosis.
Collapse
Affiliation(s)
| | - Elisabetta Rosa
- Department of Pharmacy and Research Centre on Bioactive Peptides (CIRPeB), University of Naples "Federico II", 80131 Naples, Italy
| | - Enrico Gallo
- IRCCS Synlab SDN, Via Gianturco 113, 80143 Naples, Italy
| | - Carlo Diaferia
- Department of Pharmacy and Research Centre on Bioactive Peptides (CIRPeB), University of Naples "Federico II", 80131 Naples, Italy
| | - Giancarlo Morelli
- Department of Pharmacy and Research Centre on Bioactive Peptides (CIRPeB), University of Naples "Federico II", 80131 Naples, Italy
| | - Mariano Stornaiuolo
- Department of Pharmacy and Research Centre on Bioactive Peptides (CIRPeB), University of Naples "Federico II", 80131 Naples, Italy
| | - Antonella Accardo
- Department of Pharmacy and Research Centre on Bioactive Peptides (CIRPeB), University of Naples "Federico II", 80131 Naples, Italy
| |
Collapse
|
175
|
Takeda A, Tachibana A, Nagumo H, Sakai-Kato K. An in vitro lipid-mixing assay to investigate the fusion between small extracellular vesicles and endosome. Anal Biochem 2023; 669:115130. [PMID: 36963556 DOI: 10.1016/j.ab.2023.115130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/06/2023] [Accepted: 03/21/2023] [Indexed: 03/26/2023]
Abstract
Small extracellular vesicles (sEVs) such as exosomes can efficiently deliver nucleic acids into the cytosol of recipient cells. However, the molecular mechanism of the subsequent fusion with an endosome is not well understood. In this study, we developed an in vitro lipid-mixing assay using an endosomal-mimicking anionic liposome to investigate the fusion between sEVs and endosomes. We observed that the particle number ratio between the sEVs and the anionic liposomes, the diameter of the liposomes, and the buffer pH were all important for fusion activity. Furthermore, we optimized the liposomal lipid composition and demonstrated that incorporating the anionic lipid bis(monooleoylglycero) phosphate and cholesterol was important for efficient and reliable fusion. Our in vitro assay suggested that a decrease in pH increased the fusion activity. Additionally, it was suggested that this pH-dependent increase in the fusion activity was predominantly due to a change in the sEVs. sEVs possess a larger fusion activity than artificial liposomes that mimic the physicochemical properties of exosomes. These results are consistent with those of previous in vivo studies, supporting the physiological relevance of our system. This study provides an important platform for further research to clarify the molecular mechanisms of fusion between sEVs and endosomes.
Collapse
Affiliation(s)
- Arisa Takeda
- School of Pharmacy, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo, 108-8641, Japan
| | - Asuka Tachibana
- School of Pharmacy, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo, 108-8641, Japan
| | - Hiroki Nagumo
- School of Pharmacy, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo, 108-8641, Japan
| | - Kumiko Sakai-Kato
- School of Pharmacy, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo, 108-8641, Japan.
| |
Collapse
|
176
|
Yao Y, Ko Y, Grasman G, Raymond JE, Lahann J. The steep road to nonviral nanomedicines: Frequent challenges and culprits in designing nanoparticles for gene therapy. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2023; 14:351-361. [PMID: 36959977 PMCID: PMC10028570 DOI: 10.3762/bjnano.14.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
The potential of therapeutically loaded nanoparticles (NPs) has been successfully demonstrated during the last decade, with NP-mediated nonviral gene delivery gathering significant attention as highlighted by the broad clinical acceptance of mRNA-based COVID-19 vaccines. A significant barrier to progress in this emerging area is the wild variability of approaches reported in published literature regarding nanoparticle characterizations. Here, we provide a brief overview of the current status and outline important concerns regarding the need for standardized protocols to evaluate NP uptake, NP transfection efficacy, drug dose determination, and variability of nonviral gene delivery systems. Based on these concerns, we propose wide adherence to multimodal, multiparameter, and multistudy analysis of NP systems. Adoption of these proposed approaches will ensure improved transparency, provide a better basis for interlaboratory comparisons, and will simplify judging the significance of new findings in a broader context, all critical requirements for advancing the field of nonviral gene delivery.
Collapse
Affiliation(s)
- Yao Yao
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yeongun Ko
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- School of Polymer Science and Engineering, Chonnam National University, Buk-gu, Gwangju 61186, South Korea
| | - Grant Grasman
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jeffery E Raymond
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Joerg Lahann
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
177
|
Ismaeel A, Van Pelt DW, Hettinger ZR, Fu X, Richards CI, Butterfield TA, Petrocelli JJ, Vechetti IJ, Confides AL, Drummond MJ, Dupont-Versteegden EE. Extracellular vesicle distribution and localization in skeletal muscle at rest and following disuse atrophy. Skelet Muscle 2023; 13:6. [PMID: 36895061 PMCID: PMC9999658 DOI: 10.1186/s13395-023-00315-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 02/24/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Skeletal muscle (SkM) is a large, secretory organ that produces and releases myokines that can have autocrine, paracrine, and endocrine effects. Whether extracellular vesicles (EVs) also play a role in the SkM adaptive response and ability to communicate with other tissues is not well understood. The purpose of this study was to investigate EV biogenesis factors, marker expression, and localization across cell types in the skeletal muscle. We also aimed to investigate whether EV concentrations are altered by disuse atrophy. METHODS To identify the potential markers of SkM-derived EVs, EVs were isolated from rat serum using density gradient ultracentrifugation, followed by fluorescence correlation spectroscopy measurements or qPCR. Single-cell RNA sequencing (scRNA-seq) data from rat SkM were analyzed to assess the EV biogenesis factor expression, and cellular localization of tetraspanins was investigated by immunohistochemistry. Finally, to assess the effects of mechanical unloading on EV expression in vivo, EV concentrations were measured in the serum by nanoparticle tracking analysis in both a rat and human model of disuse. RESULTS In this study, we show that the widely used markers of SkM-derived EVs, α-sarcoglycan and miR-1, are undetectable in serum EVs. We also found that EV biogenesis factors, including the tetraspanins CD63, CD9, and CD81, are expressed by a variety of cell types in SkM. SkM sections showed very low detection of CD63, CD9, and CD81 in myofibers and instead accumulation within the interstitial space. Furthermore, although there were no differences in serum EV concentrations following hindlimb suspension in rats, serum EV concentrations were elevated in human subjects after bed rest. CONCLUSIONS Our findings provide insight into the distribution and localization of EVs in SkM and demonstrate the importance of methodological guidelines in SkM EV research.
Collapse
Affiliation(s)
- Ahmed Ismaeel
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Douglas W Van Pelt
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physical Therapy, University of Kentucky, Lexington, USA
| | - Zachary R Hettinger
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physical Therapy, University of Kentucky, Lexington, USA
| | - Xu Fu
- Department of Chemistry, University of Kentucky, Lexington, KY, USA
| | | | - Timothy A Butterfield
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Jonathan J Petrocelli
- Department of Physical Therapy & Athletic Training, University of Utah, Salt Lake City, UT, USA
| | - Ivan J Vechetti
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Amy L Confides
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physical Therapy, University of Kentucky, Lexington, USA
| | - Micah J Drummond
- Department of Physical Therapy & Athletic Training, University of Utah, Salt Lake City, UT, USA
| | - Esther E Dupont-Versteegden
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA.
- Department of Physiology, University of Kentucky, Lexington, KY, USA.
- Department of Physical Therapy, University of Kentucky, Lexington, USA.
- College of Health Sciences, University of Kentucky, 900 S. Limestone, CTW 210E, Lexington, KY, 40536-0200, USA.
| |
Collapse
|
178
|
Spiers HVM, Stadler LKJ, Smith H, Kosmoliaptsis V. Extracellular Vesicles as Drug Delivery Systems in Organ Transplantation: The Next Frontier. Pharmaceutics 2023; 15:891. [PMID: 36986753 PMCID: PMC10052210 DOI: 10.3390/pharmaceutics15030891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/12/2023] Open
Abstract
Extracellular vesicles are lipid bilayer-delimited nanoparticles excreted into the extracellular space by all cells. They carry a cargo rich in proteins, lipids and DNA, as well as a full complement of RNA species, which they deliver to recipient cells to induce downstream signalling, and they play a key role in many physiological and pathological processes. There is evidence that native and hybrid EVs may be used as effective drug delivery systems, with their intrinsic ability to protect and deliver a functional cargo by utilising endogenous cellular mechanisms making them attractive as therapeutics. Organ transplantation is the gold standard for treatment for suitable patients with end-stage organ failure. However, significant challenges still remain in organ transplantation; prevention of graft rejection requires heavy immunosuppression and the lack of donor organs results in a failure to meet demand, as manifested by growing waiting lists. Pre-clinical studies have demonstrated the ability of EVs to prevent rejection in transplantation and mitigate ischemia reperfusion injury in several disease models. The findings of this work have made clinical translation of EVs possible, with several clinical trials actively recruiting patients. However, there is much to be uncovered, and it is essential to understand the mechanisms behind the therapeutic benefits of EVs. Machine perfusion of isolated organs provides an unparalleled platform for the investigation of EV biology and the testing of the pharmacokinetic and pharmacodynamic properties of EVs. This review classifies EVs and their biogenesis routes, and discusses the isolation and characterisation methods adopted by the international EV research community, before delving into what is known about EVs as drug delivery systems and why organ transplantation represents an ideal platform for their development as drug delivery systems.
Collapse
Affiliation(s)
- Harry V M Spiers
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
- NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Lukas K J Stadler
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
- NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Hugo Smith
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Vasilis Kosmoliaptsis
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
- NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
179
|
Sympathetic neurons secrete retrogradely transported TrkA on extracellular vesicles. Sci Rep 2023; 13:3657. [PMID: 36871060 PMCID: PMC9985603 DOI: 10.1038/s41598-023-30728-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Proper wiring of the peripheral nervous system relies on neurotrophic signaling via nerve growth factor (NGF). NGF secreted by target organs (i.e. eye) binds to the TrkA receptor expressed on the distal axons of postganglionic neurons. Upon binding, TrkA is internalized into a signaling endosome and retrogradely trafficked back to the soma and into the dendrites to promote cell survival and postsynaptic maturation, respectively. Much progress has been made in recent years to define the fate of the retrogradely trafficked TrkA signaling endosome, yet it has not been fully characterized. Here we investigate extracellular vesicles (EVs) as a novel route of neurotrophic signaling. Using the mouse superior cervical ganglion (SCG) as a model, we isolate EVs derived from sympathetic cultures and characterize them using immunoblot assays, nanoparticle tracking analysis, and cryo-electron microscopy. Furthermore, using a compartmentalized culture system, we find that TrkA derived from endosomes originating in the distal axon can be detected on EVs secreted from the somatodendritic domain. In addition, inhibition of classic TrkA downstream pathways, specifically in somatodendritic compartments, greatly decreases TrkA packaging into EVs. Our results suggest a novel trafficking route for TrkA: it can travel long distances to the cell body, be packaged into EVs, and be secreted. Secretion of TrkA via EVs appears to be regulated by its own downstream effector cascades, raising intriguing future questions about novel functionalities associated with TrkA+ EVs.
Collapse
|
180
|
Oh JY, Marques MB, Xu X, Li J, Genschmer KR, Phillips E, Chimento MF, Mobley J, Gaggar A, Patel RP. Different-sized extracellular vesicles derived from stored red blood cells package diverse cargoes and cause distinct cellular effects. Transfusion 2023; 63:586-600. [PMID: 36752125 PMCID: PMC10033430 DOI: 10.1111/trf.17271] [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: 09/06/2022] [Revised: 11/08/2022] [Accepted: 12/22/2022] [Indexed: 02/09/2023]
Abstract
BACKGROUND The formation of extracellular vesicles (EVs) occurs during cold storage of RBCs. Transfusion of EVs may contribute to adverse responses in recipients receiving RBCs. However, EVs are poorly characterized with limited data on whether distinct vesicles are formed, their composition, and potential biological effects. STUDY DESIGN AND METHODS Stored RBC-derived EVs were purified using protocols that separate larger microvesicle-like EVs (LEVs) from smaller exosome-like vesicles (SEVs). Vesicles were analyzed by electron microscopy, content of hemoglobin, heme, and proteins (by mass spectrometry), and the potential to mediate lipid peroxidation and endothelial cell permeability in vitro. RESULTS SEVs were characterized by having an electron-dense double membrane whereas LEVs had more uniform electron density across the particles. No differences in hemoglobin nor heme levels per particle were observed, however, due to smaller volumes, SEVs had higher concentrations of oxyHb and heme. Both particles contained antioxidant proteins peroxiredoxin-2 and copper/zinc superoxide dismutase, these were present in higher molecular weight fractions in SEVs suggesting either oxidized proteins are preferentially packaged into smaller vesicles and/or that the environment associated with SEVs is more pro-oxidative. Furthermore, total glutathione (GSH + GSSG) levels were lower in SEVs. Both EVs mediated oxidation of liposomes that were prevented by hemopexin, identifying heme as the pro-oxidant effector. Addition of SEVs, but not LEVs, induced endothelial permeability in a process also prevented by hemopexin. CONCLUSION These data show that distinct EVs are formed during cold storage of RBCs with smaller particles being more likely to mediate pro-oxidant and inflammatory effects associated with heme.
Collapse
Affiliation(s)
- Joo-Yeun Oh
- Department of Pathology, University of Alabama at Birmingham
| | | | - Xin Xu
- Department of Medicine, University of Alabama at Birmingham
- Department of Program in Protease and Matrix Biology, University of Alabama at Birmingham
| | - Jindong Li
- Department of Medicine, University of Alabama at Birmingham
- Department of Program in Protease and Matrix Biology, University of Alabama at Birmingham
| | | | - Edward Phillips
- Department of High Resolution Imaging Shared Facility, University of Alabama at Birmingham
| | - Melissa F. Chimento
- Department of High Resolution Imaging Shared Facility, University of Alabama at Birmingham
| | - James Mobley
- Department of Anesthesiolgy, University of Alabama at Birmingham
| | - Amit Gaggar
- Department of Medicine, University of Alabama at Birmingham
- Department of Program in Protease and Matrix Biology, University of Alabama at Birmingham
| | - Rakesh P. Patel
- Department of Pathology, University of Alabama at Birmingham
- Department of Center for Free Radical Biology, University of Alabama at Birmingham
| |
Collapse
|
181
|
Hettiarachchi S, Cha H, Ouyang L, Mudugamuwa A, An H, Kijanka G, Kashaninejad N, Nguyen NT, Zhang J. Recent microfluidic advances in submicron to nanoparticle manipulation and separation. LAB ON A CHIP 2023; 23:982-1010. [PMID: 36367456 DOI: 10.1039/d2lc00793b] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Manipulation and separation of submicron and nanoparticles are indispensable in many chemical, biological, medical, and environmental applications. Conventional technologies such as ultracentrifugation, ultrafiltration, size exclusion chromatography, precipitation and immunoaffinity capture are limited by high cost, low resolution, low purity or the risk of damage to biological particles. Microfluidics can accurately control fluid flow in channels with dimensions of tens of micrometres. Rapid microfluidics advancement has enabled precise sorting and isolating of nanoparticles with better resolution and efficiency than conventional technologies. This paper comprehensively studies the latest progress in microfluidic technology for submicron and nanoparticle manipulation. We first summarise the principles of the traditional techniques for manipulating nanoparticles. Following the classification of microfluidic techniques as active, passive, and hybrid approaches, we elaborate on the physics, device design, working mechanism and applications of each technique. We also compare the merits and demerits of different microfluidic techniques and benchmark them with conventional technologies. Concurrently, we summarise seven standard post-separation detection techniques for nanoparticles. Finally, we discuss current challenges and future perspectives on microfluidic technology for nanoparticle manipulation and separation.
Collapse
Affiliation(s)
- Samith Hettiarachchi
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Haotian Cha
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Lingxi Ouyang
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | | | - Hongjie An
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Gregor Kijanka
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Navid Kashaninejad
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Jun Zhang
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| |
Collapse
|
182
|
Sharma V, Nikolajeff F, Kumar S. Employing nanoparticle tracking analysis of salivary neuronal exosomes for early detection of neurodegenerative diseases. Transl Neurodegener 2023; 12:7. [PMID: 36747288 PMCID: PMC9903484 DOI: 10.1186/s40035-023-00339-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/23/2023] [Indexed: 02/08/2023] Open
Abstract
Neurodegenerative diseases are a set of progressive and currently incurable diseases that are primarily caused by neuron degeneration. Neurodegenerative diseases often lead to cognitive impairment and dyskinesias. It is now well recognized that molecular events precede the onset of clinical symptoms by years. Over the past decade, intensive research attempts have been aimed at the early diagnosis of these diseases. Recently, exosomes have been shown to play a pivotal role in the occurrence and progression of many diseases including cancer and neurodegenerative diseases. Additionally, because exosomes can cross the blood-brain barrier, they may serve as a diagnostic tool for neural dysfunction. In this review, we detail the mechanisms and current challenges of these diseases, briefly review the role of exosomes in the progression of neurodegenerative diseases, and propose a novel strategy based on salivary neuronal exosomes and nanoparticle tracking analysis that could be employed for screening the early onset of neurodegenerative diseases.
Collapse
Affiliation(s)
- Vaibhav Sharma
- Department of Health, Education and Technology, Lulea University of Technology, Lulea, Sweden.
| | - Fredrik Nikolajeff
- Department of Health, Education and Technology, Lulea University of Technology, Lulea, Sweden
| | - Saroj Kumar
- Department of Health, Education and Technology, Lulea University of Technology, Lulea, Sweden.
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
183
|
Yin T, Liu Y, Ji W, Zhuang J, Chen X, Gong B, Chu J, Liang W, Gao J, Yin Y. Engineered mesenchymal stem cell-derived extracellular vesicles: A state-of-the-art multifunctional weapon against Alzheimer's disease. Theranostics 2023; 13:1264-1285. [PMID: 36923533 PMCID: PMC10008732 DOI: 10.7150/thno.81860] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/21/2023] [Indexed: 02/15/2023] Open
Abstract
With the increase of population aging, the number of Alzheimer's disease (AD) patients is also increasing. According to current estimates, approximately 11% of people over 65 suffer from AD, and that percentage rises to 42% among people over 85. However, no effective treatment capable of decelerating or stopping AD progression is available. Furthermore, AD-targeted drugs composed of synthetic molecules pose concerns regarding biodegradation, clearance, immune response, and neurotoxicity. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) are essential intercellular communication mediators holding great promise as AD therapeutics owing to their biocompatibility, versatility, effortless storage, superior safety, and the ability to transport messenger and noncoding RNAs, proteins, lipids, DNAs, and other bioactive compounds derived from cells. The functionalisation and engineering strategies of MSC-EVs are highlighted (e.g. preconditioning, drug loading, surface modification, and artificial EV fabrication), which could improve AD treatment by multiple therapeutic effects, including clearing abnormal protein accumulation and achieving neuroprotection and immunomodulatory effects. Herein, this review summarises state-of-the-art strategies to engineer MSC-EVs, discusses progress in their use as AD therapeutics, presents the perspectives and challenges associated with the related clinical applications, and concludes that engineered MSC-EVs show immense potential in AD therapy.
Collapse
Affiliation(s)
- Tong Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Yan Liu
- Department of Clinical Pharmacy, Xinhua Hospital; Clinical pharmacy innovation institute, Shanghai Jiao Tong University of Medicine, Shanghai 200000, China
| | - Wenbo Ji
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Jianhua Zhuang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Xiaohan Chen
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Baofeng Gong
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Jianjian Chu
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Wendanqi Liang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - You Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai 200003, China
| |
Collapse
|
184
|
Sausset R, Krupova Z, Guédon E, Peron S, Grangier A, Petit M, De Sordi L, De Paepe M. Comparison of interferometric light microscopy with nanoparticle tracking analysis for the study of extracellular vesicles and bacteriophages. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e75. [PMID: 38938523 PMCID: PMC11080698 DOI: 10.1002/jex2.75] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 06/29/2024]
Abstract
Research on extracellular vesicles (EVs) and bacteriophages (phages) has been steadily expanding over the past decades as many of their roles in medicine, biology, and ecosystems have been unveiled. Such interest has brought about the need for new tools to quantify and determine the sizes of these biological nanoparticles. A new device based on interferometric light microscopy (ILM), the Videodrop, was recently developed for this purpose. Here, we compared this new device to two nanoparticle tracking analysis (NTA) devices, the NanoSight and the ZetaView, for the analysis of EVs and phages. We used EVs isolated from bacteria, fecal samples, bovine milk and human cells, and phages of various sizes and shape, ranging from 30 to 120 nm of diameter. While NTA instruments correctly enumerated most phages, the Videodrop detected only the largest one, indicating a lower sensitivity threshold compared to the NTA devices. Nevertheless, the performance of the Videodrop compared favourably to that of the NTA devices for the determination of the concentration of eukaryotic EV samples. The NanoSight instrument provided the most precise size distributions but the Videodrop was by far the most time-saving device, making it worthy of consideration for studies conducted on a large number of samples composed of nanoparticles larger than 90 nm.
Collapse
Affiliation(s)
- Romain Sausset
- Micalis Institute, INRAE, AgroParisTechUniversité Paris‐SaclayJouy‐en‐JosasFrance
- Myriade68 boulevard de Port RoyalParisFrance
- Centre de Recherche Saint AntoineSorbonne Université, INSERMParisFrance
| | - Zuzana Krupova
- Excilone, Departement R&D6 rue Blaise Pascal, Parc Euclide, Bat. AElancourtFrance
| | | | | | - Alice Grangier
- Laboratoire MSC Matière et Systèmes ComplexesCNRS UMR 7057Université Paris CitéParisFrance
| | - Marie‐Agnès Petit
- Micalis Institute, INRAE, AgroParisTechUniversité Paris‐SaclayJouy‐en‐JosasFrance
| | - Luisa De Sordi
- Centre de Recherche Saint AntoineSorbonne Université, INSERMParisFrance
| | - Marianne De Paepe
- Micalis Institute, INRAE, AgroParisTechUniversité Paris‐SaclayJouy‐en‐JosasFrance
| |
Collapse
|
185
|
Ye S, You Q, Song S, Wang H, Wang C, Zhu L, Yang Y. Nanostructures and Nanotechnologies for the Detection of Extracellular Vesicle. Adv Biol (Weinh) 2023; 7:e2200201. [PMID: 36394211 DOI: 10.1002/adbi.202200201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/17/2022] [Indexed: 11/19/2022]
Abstract
Liquid biopsy has been taken as a minimally invasive examination and a promising surrogate to the clinically applied tissue-based test for the diagnosis and molecular analysis of cancer. Extracellular vesicles (EVs) carry complex molecular information from the tumor, allowing for the multicomponent analysis of cancer and would be beneficial to personalized medicine. In this review, the advanced nanomaterials and nanotechniques for the detection and molecular profiling of EVs, highlight the advantages of nanotechnology in the high-purity isolation and the high-sensitive and high-specific identification of EVs, are summarized. An outlook on the clinical application of nanotechnology-based liquid biopsy in the diagnosis, prognostication, and surveillance of cancer is also provided. It provides information for developing liquid biopsy based on EVs by discussing the advantages and challenges of functionalized nanomaterials and various nanotechnologies.
Collapse
Affiliation(s)
- Siyuan Ye
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China.,Department of Chemistry, Tsinghua University, Beijing, 100084, P. R. China
| | - Qing You
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Shuya Song
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Huayi Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China.,Translational Medicine Center, Chinese Institute for Brain Research (CIBR), Beijing, 102206, P. R. China
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
186
|
Stem Cell-derived Extracellular Vesicles: A Promising Nano Delivery Platform to the Brain? Stem Cell Rev Rep 2023; 19:285-308. [PMID: 36173500 DOI: 10.1007/s12015-022-10455-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2022] [Indexed: 02/07/2023]
Abstract
A very important cause of the frustration with drug therapy for central nervous system (CNS) diseases is the failure of drug delivery. The blood-brain barrier (BBB) prevents most therapeutic molecules from entering the brain while maintaining CNS homeostasis. Scientists are keen to develop new brain drug delivery systems to solve this dilemma. Extracellular vesicles (EVs), as a class of naturally derived nanoscale vesicles, have been extensively studied in drug delivery due to their superior properties. This review will briefly present current brain drug delivery strategies, including invasive and non-invasive techniques that target the brain, and the application of nanocarriers developed for brain drug delivery in recent years, especially EVs. The cellular origin of EVs affects the surface protein, size, yield, luminal composition, and other properties of EVs, which are also crucial in determining whether EVs are useful as drug carriers. Stem cell-derived EVs, which inherit the properties of parental cells and avoid the drawbacks of cell therapy, have always been favored by researchers. Thus, in this review, we will focus on the application of stem cell-derived EVs for drug delivery in the CNS. Various nucleic acids, proteins, and small-molecule drugs are loaded into EVs with or without modification and undergo targeted delivery to the brain to achieve their therapeutic effects. In addition, the challenges facing the clinical application of EVs as drug carriers will also be discussed. The directions of future efforts may be to improve drug loading efficiency and precise targeting.
Collapse
|
187
|
Schiller EA, Cohen K, Lin X, El-Khawam R, Hanna N. Extracellular Vesicle-microRNAs as Diagnostic Biomarkers in Preterm Neonates. Int J Mol Sci 2023; 24:2622. [PMID: 36768944 PMCID: PMC9916767 DOI: 10.3390/ijms24032622] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Neonates born prematurely (<37 weeks of gestation) are at a significantly increased risk of developing inflammatory conditions associated with high mortality rates, including necrotizing enterocolitis, bronchopulmonary dysplasia, and hypoxic-ischemic brain damage. Recently, research has focused on characterizing the content of extracellular vesicles (EVs), particularly microRNAs (miRNAs), for diagnostic use. Here, we describe the most recent work on EVs-miRNAs biomarkers discovery for conditions that commonly affect premature neonates.
Collapse
Affiliation(s)
- Emily A. Schiller
- Department of Foundational Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, USA
| | - Koral Cohen
- Department of Foundational Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, USA
| | - Xinhua Lin
- Department of Foundational Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, USA
| | - Rania El-Khawam
- Department of Pediatrics, Division of Neonatology, New York University Langone Long Island Hospital, Mineola, NY 11501, USA
| | - Nazeeh Hanna
- Department of Foundational Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, USA
- Department of Pediatrics, Division of Neonatology, New York University Langone Long Island Hospital, Mineola, NY 11501, USA
| |
Collapse
|
188
|
Biagiotti S, Abbas F, Montanari M, Barattini C, Rossi L, Magnani M, Papa S, Canonico B. Extracellular Vesicles as New Players in Drug Delivery: A Focus on Red Blood Cells-Derived EVs. Pharmaceutics 2023; 15:365. [PMID: 36839687 PMCID: PMC9961903 DOI: 10.3390/pharmaceutics15020365] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
The article is divided into several sections, focusing on extracellular vesicles' (EVs) nature, features, commonly employed methodologies and strategies for their isolation/preparation, and their characterization/visualization. This work aims to give an overview of advances in EVs' extensive nanomedical-drug delivery applications. Furthermore, considerations for EVs translation to clinical application are summarized here, before focusing the review on a special kind of extracellular vesicles, the ones derived from red blood cells (RBCEVs). Generally, employing EVs as drug carriers means managing entities with advantageous properties over synthetic vehicles or nanoparticles. Besides the fact that certain EVs also reveal intrinsic therapeutic characteristics, in regenerative medicine, EVs nanosize, lipidomic and proteomic profiles enable them to pass biologic barriers and display cell/tissue tropisms; indeed, EVs engineering can further optimize their organ targeting. In the second part of the review, we focus our attention on RBCEVs. First, we describe the biogenesis and composition of those naturally produced by red blood cells (RBCs) under physiological and pathological conditions. Afterwards, we discuss the current procedures to isolate and/or produce RBCEVs in the lab and to load a specific cargo for therapeutic exploitation. Finally, we disclose the most recent applications of RBCEVs at the in vitro and preclinical research level and their potential industrial exploitation. In conclusion, RBCEVs can be, in the near future, a very promising and versatile platform for several clinical applications and pharmaceutical exploitations.
Collapse
Affiliation(s)
- Sara Biagiotti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy
| | - Faiza Abbas
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy
| | - Mariele Montanari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy
| | - Chiara Barattini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy
- AcZon s.r.l., 40050 Monte San Pietro, BO, Italy
| | - Luigia Rossi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy
| | - Stefano Papa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy
| | - Barbara Canonico
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy
| |
Collapse
|
189
|
Ortiz-Orruño U, Quidant R, van Hulst NF, Liebel M, Ortega Arroyo J. Simultaneous Sizing and Refractive Index Analysis of Heterogeneous Nanoparticle Suspensions. ACS NANO 2023; 17:221-229. [PMID: 36525614 PMCID: PMC9835976 DOI: 10.1021/acsnano.2c06883] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 12/13/2022] [Indexed: 05/25/2023]
Abstract
Rapid and reliable characterization of heterogeneous nanoparticle suspensions is a key technology across the nanosciences. Although approaches exist for homogeneous samples, they are often unsuitable for polydisperse suspensions, as particles of different sizes and compositions can lead to indistinguishable signals at the detector. Here, we introduce holographic nanoparticle tracking analysis, holoNTA, as a straightforward methodology that decouples size and material refractive index contributions. HoloNTA is applicable to any heterogeneous nanoparticle sample and has the sensitivity to measure the intrinsic heterogeneity of the sample. Specifically, we combined high dynamic range k-space imaging with holographic 3D single-particle tracking. This strategy enables long-term tracking by extending the imaging volume and delivers precise and accurate estimates of both scattering amplitude and diffusion coefficient of individual nanoparticles, from which particle refractive index and hydrodynamic size are determined. We specifically demonstrate, by simulations and experiments, that irrespective of localization uncertainty and size, the sizing sensitivity is improved as our extended detection volume yields considerably longer particle trajectories than previously reported by comparable technologies. As validation, we measured both homogeneous and heterogeneous suspensions of nanoparticles in the 40-250 nm size range and further monitored protein corona formation, where we identified subtle differences between the nanoparticle-protein complexes derived from avidin, bovine serum albumin, and streptavidin. We foresee that our approach will find many applications of both fundamental and applied nature where routine quantification and sizing of nanoparticles are required.
Collapse
Affiliation(s)
- Unai Ortiz-Orruño
- ICFO,
Institut de Ciencies Fotoniques, The Barcelona Institute of Science
and Technology, Castelldefels08860, Spain
| | - Romain Quidant
- Nanophotonic
Systems Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich8092, Switzerland
| | - Niek F. van Hulst
- ICFO,
Institut de Ciencies Fotoniques, The Barcelona Institute of Science
and Technology, Castelldefels08860, Spain
- ICREA,
Institució Catalana de Recerca i Estudis Avançats, Barcelona08010, Spain
| | - Matz Liebel
- ICFO,
Institut de Ciencies Fotoniques, The Barcelona Institute of Science
and Technology, Castelldefels08860, Spain
| | - Jaime Ortega Arroyo
- Nanophotonic
Systems Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich8092, Switzerland
| |
Collapse
|
190
|
Suthar J, Taub M, Carney RP, Williams GR, Guldin S. Recent developments in biosensing methods for extracellular vesicle protein characterization. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1839. [PMID: 35999185 PMCID: PMC10078591 DOI: 10.1002/wnan.1839] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/27/2022] [Accepted: 07/13/2022] [Indexed: 01/31/2023]
Abstract
Research into extracellular vesicles (EVs) has grown significantly over the last few decades with EVs being widely regarded as a source of biomarkers for human health and disease with massive clinical potential. Secreted by every cell type in the body, EVs report on the internal cellular conditions across all tissue types. Their presence in readily accessible biofluids makes the potential of EV biosensing highly attractive as a noninvasive diagnostic platform via liquid biopsies. However, their small size (50-250 nm), inherent heterogeneity, and the complexity of the native biofluids introduce challenges for effective characterization, thus, limiting their clinical utility. This has led to a surge in the development of various novel EV biosensing techniques, with capabilities beyond those of conventional methods that have been directly transferred from cell biology. In this review, key detection principles used for EV biosensing are summarized, with a focus on some of the most recent and fundamental developments in the field over the last 5 years. This article is categorized under: Diagnostic Tools > Biosensing Diagnostic Tools > In Vitro Nanoparticle-Based Sensing.
Collapse
Affiliation(s)
- Jugal Suthar
- Department of Chemical Engineering, University College London, London, UK.,UCL School of Pharmacy, University College London, London, UK
| | - Marissa Taub
- UCL School of Pharmacy, University College London, London, UK
| | - Randy P Carney
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
| | | | - Stefan Guldin
- Department of Chemical Engineering, University College London, London, UK
| |
Collapse
|
191
|
Jackson KK, Mata C, Marcus RK. A rapid capillary-channeled polymer (C-CP) fiber spin-down tip approach for the isolation of plant-derived extracellular vesicles (PDEVs) from 20 common fruit and vegetable sources. Talanta 2023; 252:123779. [DOI: 10.1016/j.talanta.2022.123779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/18/2022] [Accepted: 07/24/2022] [Indexed: 10/15/2022]
|
192
|
Abstract
This review focuses on nonlytic outer membrane vesicles (OMVs), a subtype of bacterial extracellular vesicles (BEVs) produced by Gram-negative organisms focusing on the mechanisms of their biogenesis, cargo, and function. Throughout, we highlight issues concerning the characterization of OMVs and distinguishing them from other types of BEVs. We also highlight the shortcomings of commonly used methodologies for the study of BEVs that impact the interpretation of their functionality and suggest solutions to standardize protocols for OMV studies.
Collapse
Affiliation(s)
| | - Simon R. Carding
- Quadram Institute Bioscience, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
193
|
Walker SA, Davidovich I, Yang Y, Lai A, Goncalves JP, Deliwala V, Busatto S, Shapiro S, Koifman N, Salomon C, Talmon Y, Wolfram J. Sucrose-based cryoprotective storage of extracellular vesicles. EXTRACELLULAR VESICLE 2022; 1:100016. [PMID: 38665624 PMCID: PMC11044822 DOI: 10.1016/j.vesic.2022.100016] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Advancements in extracellular vesicle (EV) studies necessitate the development of optimized storage conditions to ensure preservation of physical and biochemical characteristics. In this study, the most common buffer for EV storage (phosphate-buffered saline/PBS) was compared to a cryoprotective 5% sucrose solution. The size distribution and concentration of EVs from two different sources changed to a greater extent after -80 °C storage in PBS compared to the sucrose solution. Additionally, molecular surface protrusions and transmembrane proteins were more prevalent in EVs stored in the sucrose solution compared to those stored in PBS. This study demonstrates, for the first time, that distinct ring-like molecular complexes and cristae-like folded membranous structures are visible upon EV degradation. Taken together, the size, concentration, molecular surface extensions, and transmembrane proteins of EVs varied substantially based on the buffer used for -80 °C storage, suggesting that biocompatible cryoprotectants, such as sucrose, should be considered for EV studies.
Collapse
Affiliation(s)
- Sierra A. Walker
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Irina Davidovich
- Department of Chemical Engineering and the Russell Berrie Nanotechnology Institute (RBNI), Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Yubo Yang
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Andrew Lai
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4006, Australia
| | - Jenifer Pendiuk Goncalves
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD, Australia
| | - Vatsal Deliwala
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD, Australia
| | - Sara Busatto
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, United States
- Department of Surgery, Harvard Medical School, Boston, MA 02115, United States
| | - Shane Shapiro
- Department of Orthopedic Surgery, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Na’ama Koifman
- Center of Microscopy and Microanalysis, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4006, Australia
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago 8320000, Chile
| | - Yeshayahu Talmon
- Department of Chemical Engineering and the Russell Berrie Nanotechnology Institute (RBNI), Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Joy Wolfram
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
194
|
Dumontel B, Susa F, Limongi T, Vighetto V, Debellis D, Canta M, Cauda V. Nanotechnological engineering of extracellular vesicles for the development of actively targeted hybrid nanodevices. Cell Biosci 2022; 12:61. [PMID: 35568919 PMCID: PMC9107671 DOI: 10.1186/s13578-022-00784-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/06/2022] [Indexed: 12/18/2022] Open
Abstract
Background We propose an efficient method to modify B-cell derived EVs by loading them with a nanotherapeutic stimuli-responsive cargo and equipping them with antibodies for efficient targeting of lymphoma cells. Results The post-isolation engineering of the EVs is accomplished by a freeze–thaw method to load therapeutically-active zinc oxide nanocrystals (ZnO NCs), obtaining the so-called TrojanNanoHorse (TNH) to recall the biomimetism and cytotoxic potential of this novel nanoconstruct. TNHs are further modified at their surface with anti-CD20 monoclonal antibodies (TNHCD20) achieving specific targeting against lymphoid cancer cell line. The in vitro characterization is carried out on CD20+ lymphoid Daudi cell line, CD20-negative cancerous myeloid cells (HL60) and the healthy counterpart (B lymphocytes). The TNH shows nanosized structure, high colloidal stability, even over time, and good hemocompatibility. The in vitro characterization shows the high biocompatibility, targeting specificity and cytotoxic capability. Importantly, the selectivity of TNHCD20 demonstrates significantly higher interaction towards the target lymphoid Daudi cell line compared to the CD20-negative cancerous myeloid cells (HL60) and the healthy counterpart (lymphocytes). An enhanced cytotoxicity directed against Daudi cancer cells is demonstrated after the TNHCD20 activation with high-energy ultrasound shock-waves (SW). Conclusion This work demonstrates the efficient re-engineering of EVs, derived from healthy cells, with inorganic nanoparticles and monoclonal antibodies. The obtained hybrid nanoconstructs can be on-demand activated by an external stimulation, here acoustic pressure waves, to exploit a cytotoxic effect conveyed by the ZnO NCs cargo against selected cancer cells. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00784-9.
Collapse
|
195
|
Clément V, Roy V, Paré B, Goulet CR, Deschênes LT, Berthod F, Bolduc S, Gros-Louis F. Tridimensional cell culture of dermal fibroblasts promotes exosome-mediated secretion of extracellular matrix proteins. Sci Rep 2022; 12:19786. [PMID: 36396670 PMCID: PMC9672399 DOI: 10.1038/s41598-022-23433-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 10/31/2022] [Indexed: 11/19/2022] Open
Abstract
Extracellular matrix (ECM) secretion, deposition and assembly are part of a whole complex biological process influencing the microenvironment and other cellular behaviors. Emerging evidence is attributing a significant role to extracellular vesicles (EVs) and exosomes in a plethora of ECM-associated functions, but the role of dermal fibroblast-derived EVs in paracrine signalling is yet unclear. Herein, we investigated the effect of exosomes isolated from stimulated human dermal fibroblasts. We report that tridimensional (3D) cell culture of dermal fibroblasts promotes secretion of exosomes carrying a large quantity of proteins involved in the formation, organisation and remodelling of the ECM. In our 3D model, gene expression was highly modulated and linked to ECM, cellular migration and proliferation, as well as inflammatory response. Mass spectrometry analysis of exosomal proteins, isolated from 3D cultured fibroblast-conditioned media, revealed ECM protein enrichment, of which many were associated with the matrisome. We also show that the cytokine interleukin 6 (IL-6) is predicted to be central to the signalling pathways related to ECM formation and contributing to cell migration and proliferation. Overall, our data suggest that dermal fibroblast-derived EVs participate in many steps of the establishment of dermis's ECM.
Collapse
Affiliation(s)
- Vincent Clément
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, Laval University Experimental Organogenesis Research Center/LOEX, CHU de Québec Research Center – Enfant-Jésus Hospital, Québec, QC Canada
| | - Vincent Roy
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, Laval University Experimental Organogenesis Research Center/LOEX, CHU de Québec Research Center – Enfant-Jésus Hospital, Québec, QC Canada
| | - Bastien Paré
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, Laval University Experimental Organogenesis Research Center/LOEX, CHU de Québec Research Center – Enfant-Jésus Hospital, Québec, QC Canada
| | - Cassandra R. Goulet
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, Laval University Experimental Organogenesis Research Center/LOEX, CHU de Québec Research Center – Enfant-Jésus Hospital, Québec, QC Canada
| | - Lydia Touzel Deschênes
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, Laval University Experimental Organogenesis Research Center/LOEX, CHU de Québec Research Center – Enfant-Jésus Hospital, Québec, QC Canada
| | - François Berthod
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, Laval University Experimental Organogenesis Research Center/LOEX, CHU de Québec Research Center – Enfant-Jésus Hospital, Québec, QC Canada
| | - Stéphane Bolduc
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, Laval University Experimental Organogenesis Research Center/LOEX, CHU de Québec Research Center – Enfant-Jésus Hospital, Québec, QC Canada
| | - François Gros-Louis
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, Laval University Experimental Organogenesis Research Center/LOEX, CHU de Québec Research Center – Enfant-Jésus Hospital, Québec, QC Canada
| |
Collapse
|
196
|
Xu J, Yang KC, Go NE, Colborne S, Ho CJ, Hosseini-Beheshti E, Lystad AH, Simonsen A, Guns ET, Morin GB, Gorski SM. Chloroquine treatment induces secretion of autophagy-related proteins and inclusion of Atg8-family proteins in distinct extracellular vesicle populations. Autophagy 2022; 18:2547-2560. [PMID: 35220892 PMCID: PMC9629075 DOI: 10.1080/15548627.2022.2039535] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chloroquine (CQ), a lysosomotropic agent, is commonly used to inhibit lysosomal degradation and macroautophagy/autophagy. Here we investigated the cell-extrinsic effects of CQ on secretion. We showed that lysosomal and autophagy inhibition by CQ altered the secretome, and induced the release of Atg8 orthologs and autophagy receptors. Atg8-family proteins, in particular, were secreted inside small extracellular vesicles (sEVs) in a lipidation-dependent manner. CQ treatment enhanced the release of Atg8-family proteins inside sEVs. Using full-length ATG16L1 and an ATG16L1 mutant that enables Atg8-family protein lipidation on double but not on single membranes, we demonstrated that LC3B is released in two distinct sEV populations: one enriched with SDCBP/Syntenin-1, CD63, and endosomal lipidated LC3B, and another that contains LC3B but is not enriched with SDCBP/Syntenin-1 or CD63, and which our data supports as originating from a double-membrane source. Our findings underscore the context-dependency of sEV heterogeneity and composition, and illustrate the integration of autophagy and sEV composition in response to lysosomal inhibition.Abbreviations: ACTB: actin beta; ANOVA: analysis of variance; ATG4B: autophagy related 4B cysteine peptidase; Atg8: autophagy related 8; ATG16L1: autophagy related 16 like 1; ATP5F1A/ATP5a: ATP synthase F1 subunit alpha; CALCOCO2: calcium binding and coiled-coil domain 2; CASP3: caspase 3; CASP7: caspase 7; CQ: chloroquine; CD9: CD9 molecule; CD63: CD63 molecule; DAPI: 4',6-diamidino-2-phenylindole; DQ-BSA: dye quenched-bovine serum albumin; ER: endoplasmic reticulum; ERN1/IRE1a: endoplasmic reticulum to nucleus signaling 1; EV: extracellular vesicles; FBS: fetal bovine serum; FDR: false discovery rate; GABARAP: GABA type A receptor-associated protein; GABARAPL2: GABA type A receptor associated protein like 2; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; GO: gene ontology; HCQ: hydroxychloroquine; HSP90AA1: heat shock protein 90 alpha family class A member 1; IP: immunoprecipitation; KO: knockout; LAMP2: lysosomal associated membrane protein 2; LIR: LC3-interacting region; LMNA: lamin A/C; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MS: mass spectrometry; NBR1: NBR1 autophagy cargo receptor; NCOA4: nuclear receptor coactivator 4; NTA: nanoparticle tracking analysis; PE: phosphatidylethanolamine; PECA: probe-level expression change averaging; SDCBP/syntenin-1: syndecan binding protein; SD: standard deviation; SE: secreted; sEV: small extracellular vesicles; SQSTM1/p62: sequestosome 1; TAX1BP1: Tax1 binding protein 1; TEM: transmission electron microscopy; TMT: tandem-mass tag; TSG101: tumor susceptibility 101; ULK1: unc-51 like autophagy activating kinase 1; WC: whole cell.
Collapse
Affiliation(s)
- Jing Xu
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada,Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Kevin C Yang
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada,Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Nancy Erro Go
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada,Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Shane Colborne
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Cally J Ho
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada,Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Elham Hosseini-Beheshti
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney NSWAustralia
| | - Alf H Lystad
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway,Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway,Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Norway
| | - Emma Tomlinson Guns
- The Vancouver Prostate Centre, Vancouver, BC, Canada,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Gregg B Morin
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Sharon M Gorski
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada,Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada,Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada,CONTACT Sharon M Gorski Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| |
Collapse
|
197
|
Vandendriessche C, Kapogiannis D, Vandenbroucke RE. Biomarker and therapeutic potential of peripheral extracellular vesicles in Alzheimer's disease. Adv Drug Deliv Rev 2022; 190:114486. [PMID: 35952829 PMCID: PMC9985115 DOI: 10.1016/j.addr.2022.114486] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/01/2022] [Accepted: 08/03/2022] [Indexed: 01/24/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived nanoparticles with an important role in intercellular communication, even across brain barriers. The bidirectional brain-barrier crossing capacity of EVs is supported by research identifying neuronal markers in peripheral EVs, as well as the brain delivery of peripherally administered EVs. In addition, EVs are reflective of their cellular origin, underlining their biomarker and therapeutic potential when released by diseased and regenerative cells, respectively. Both characteristics are of interest in Alzheimer's disease (AD) where the current biomarker profile is solely based on brain-centered readouts and effective therapeutic options are lacking. In this review, we elaborate on the role of peripheral EVs in AD. We focus on bulk EVs and specific EV subpopulations including bacterial EVs (bEVs) and neuronal-derived EVs (nEVs), which have mainly been studied from a biomarker perspective. Furthermore, we highlight the therapeutic potential of peripherally administered EVs whereby research has centered around stem cell derived EVs.
Collapse
Affiliation(s)
- Charysse Vandendriessche
- VIB Center for Inflammation Research, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Dimitrios Kapogiannis
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, USA; Department of Neurology, Johns Hopkins School of Medicine, Baltimore, USA
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
198
|
Pszczółkowska B, Olejarz W, Filipek M, Tartas A, Kubiak-Tomaszewska G, Żołnierzak A, Życieńska K, Ginter J, Lorenc T, Brzozowska B. Exosome secretion and cellular response of DU145 and PC3 after exposure to alpha radiation. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2022; 61:639-650. [PMID: 36098819 PMCID: PMC9630248 DOI: 10.1007/s00411-022-00991-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 08/20/2022] [Indexed: 06/15/2023]
Abstract
Exosomes are spherical membrane nanovesicles secreted from cells, and they play an important role in tumor immune response, metastasis, angiogenesis, and survival. Studies investigating exosomes isolated from cells exposed to photon radiation commonly used in conventional radiotherapy demonstrate the influence of this type of radiation on exosome characteristics and secretion. There is currently no research investigating the effects of densely ionizing particles such as protons and alpha radiation on exosomes. Thus we have evaluated the cellular response of human prostate cancer cells exposed to 0, 2, and 6 Gy of alpha radiation emitted from the Am-241 source. Irradiated PC3 and DU145 cell lines, characterized by differences in radiosensitivity, were studied using apoptosis, LDH, and IL-6 assays. Additionally, the corresponding concentration and size of isolated exosomes were measured using NTA. We found that exposure to ionizing radiation resulted in gross changes in viability and cell damage. There were increased amounts of apoptotic or necrotic cells as a function of radiation dose. We demonstrated that irradiated PC3 cells secrete higher quantities of exosomes compared to DU145 cells. Additionally, we also found no statistical difference in exosome size for control and irradiated cells.
Collapse
Affiliation(s)
- Beata Pszczółkowska
- Biomedical Physics Division, Faculty of Physics, University of Warsaw, 5 Pasteura Street, Warsaw, 02-093 Poland
| | - Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Medical University of Warsaw, 1 Banacha Street, Warsaw, 02-097 Poland
- Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1 Banacha Street, Warsaw, 02-097 Poland
| | - Mateusz Filipek
- Biomedical Physics Division, Faculty of Physics, University of Warsaw, 5 Pasteura Street, Warsaw, 02-093 Poland
| | - Adrianna Tartas
- Biomedical Physics Division, Faculty of Physics, University of Warsaw, 5 Pasteura Street, Warsaw, 02-093 Poland
| | - Grażyna Kubiak-Tomaszewska
- Department of Biochemistry and Pharmacogenomics, Medical University of Warsaw, 1 Banacha Street, Warsaw, 02-097 Poland
- Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1 Banacha Street, Warsaw, 02-097 Poland
| | - Aleksandra Żołnierzak
- Department of Biochemistry and Pharmacogenomics, Medical University of Warsaw, 1 Banacha Street, Warsaw, 02-097 Poland
- Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1 Banacha Street, Warsaw, 02-097 Poland
| | - Katarzyna Życieńska
- Biomedical Physics Division, Faculty of Physics, University of Warsaw, 5 Pasteura Street, Warsaw, 02-093 Poland
| | - Józef Ginter
- Biomedical Physics Division, Faculty of Physics, University of Warsaw, 5 Pasteura Street, Warsaw, 02-093 Poland
| | - Tomasz Lorenc
- 1st Department of Clinical Radiology, Medical University of Warsaw, 5 Chałubińskiego Street, Warsaw, 02-004 Poland
| | - Beata Brzozowska
- Biomedical Physics Division, Faculty of Physics, University of Warsaw, 5 Pasteura Street, Warsaw, 02-093 Poland
| |
Collapse
|
199
|
Li Z, Zhang J, Huang Y, Zhai J, Liao G, Wang Z, Ning C. Development of electroactive materials-based immunosensor towards early-stage cancer detection. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
200
|
Gao Z, Hutchins Z, Li Z, Zhong W. Offline Coupling of Asymmetrical Flow Field-Flow Fractionation and Capillary Electrophoresis for Separation of Extracellular Vesicles. Anal Chem 2022; 94:14083-14091. [PMID: 36191238 PMCID: PMC9988405 DOI: 10.1021/acs.analchem.2c03550] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Extracellular vesicles (EVs) play important roles in cell-to-cell communications and carry high potential as markers targeted in disease diagnosis, prognosis, and therapeutic development. The main obstacles to EV study are their high heterogeneity; low amounts present in samples; and physical similarity to the abundant, interfering matrix components. Multiple rounds of separation and purification are often needed prior to EV characterization and function assessment. Herein, we report the offline coupling of asymmetrical flow field-flow fractionation (AF4) and capillary electrophoresis (CE) for EV analysis. While AF4 provides gentle and fast EV separation by size, CE resolves EVs from contaminants with similar sizes but different surface charges. Employing Western Blotting, ELISA, and SEM, we confirmed that intact EVs were eluted within a stable time window under the optimal AF4 and CE conditions. We also proved that EVs could be resolved from free proteins and high-density lipoproteins by AF4 and be further separated from the low-density lipoproteins co-eluted in AF4. The effectiveness of the coupled AF4-CE system in EV analysis was demonstrated by monitoring the changes in EV secretion from cells and by direct injection of human serum and detection of serum EVs. We believe that coupling AF4 and CE can provide rapid EV quantification in biological samples with much reduced matrix interference and be valuable for the study of total EVs and EV subpopulations produced by cells or present in clinical samples.
Collapse
Affiliation(s)
- Ziting Gao
- Department of Chemistry, University of California─Riverside, Riverside, California 92521, United States
| | - Zachary Hutchins
- Department of Chemistry, University of California─Riverside, Riverside, California 92521, United States
| | - Zongbo Li
- Department of Chemistry, University of California─Riverside, Riverside, California 92521, United States
| | - Wenwan Zhong
- Department of Chemistry, University of California─Riverside, Riverside, California 92521, United States
| |
Collapse
|