1
|
Liu W, Yang H, Liu X, Cai H, Bao Y, Jiang Y, Zhou W, Yuan J, Zhang Z, Fang X. Ultrasensitive Quantification of microRNA Copy Number in Individual Extracellular Vesicles Using DNA Tetrahedron-Based Single-Molecule Imaging. Anal Chem 2025; 97:4233-4240. [PMID: 39936597 DOI: 10.1021/acs.analchem.4c07068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
The ultrasensitive detection of microRNAs (miRNAs) in extracellular vesicles (EVs) can accurately reflect the progress and metastasis of miRNA-mediated intercellular communication, providing an unprecedented opportunity for liquid biopsy. However, due to the low abundance and high heterogeneity of miRNAs in EVs, the ultrasensitive quantification and establishment of a distribution model for miRNA within native EVs remain challenging. Here, we have developed a DNA tetrahedron-based single-molecule fluorescence imaging strategy to overcome this challenge. The internalization efficiency of the probe was as high as 70% without disrupting the native structure of EVs, and combined with single-molecule fluorescence imaging, we achieved in situ imaging analysis of single-copy miRNA in individual EVs without amplification for the first time. A new distribution model for miRNAs has been revealed by statistical analysis of the copy number of miRNAs in EVs across multiple cell lines, characterized by low occupancy and a heterogeneous distribution. More importantly, we found that drug resistance cancer cells promote an increase in the number of drug resistance-related miRNAs within EVs without a corresponding increase in the number of EVs secreted, providing new insights into the EV miRNA sorting mechanisms. We anticipate that this technology will rapidly advance miRNA-mediated intercellular communication based on EVs.
Collapse
Affiliation(s)
- Weifeng Liu
- Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongwei Yang
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaolong Liu
- Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Heqi Cai
- Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuting Bao
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yifei Jiang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Wei Zhou
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Jinghe Yuan
- Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Zhen Zhang
- Huairou Research Center, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaohong Fang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
2
|
Yang Y, Metem P, Khaksaran MH, Sahu SS, Stridfeldt F, Görgens A, Zhang SL, Dev A. Plasmon-Enhanced Fluorescence of Single Extracellular Vesicles Captured in Arrayed Aluminum Nanoholes. ACS OMEGA 2024; 9:51022-51030. [PMID: 39758645 PMCID: PMC11696387 DOI: 10.1021/acsomega.4c05492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/18/2024] [Accepted: 12/02/2024] [Indexed: 01/07/2025]
Abstract
Extracellular vesicles (EVs) are nanoparticles encapsulated with a lipid bilayer, and they constitute an excellent source of biomarkers for multiple diseases. However, the heterogeneity in their molecular compositions constitutes a major challenge for their recognition and profiling, thereby limiting their application as an effective biomarker. A single-EV analysis technique is crucial to both the discovery and the detection of EV subpopulations that carry disease-specific signatures. Herein, a plasmonic nanohole array is designed for capturing single EVs and subsequently performing fluorescence detection of their membrane proteins by exploiting plasmonic amplification of the fluorescence signal. Unlike other reported methods, our design relies on an exclusive detection of single EVs captured inside nanoholes, thus allowing us to study only plasmonic effects and avoid other metal-induced phenomena while leveraging on the proximity of emitters to the plasmonic hotspots. The method is optimized through numerical simulations and verified by a combination of atomic force, scanning electron microscopy, and fluorescence microscopy. Fluorescence enhancement is then estimated by measuring the CD9 expression of small EVs derived from the human embryonic kidney (HEK293) cell line and carefully considering the spatial distribution of emission and excitation intensities. Fluorescence intensities of immunostained EVs show a moderate overall enhancement of intensity and follow the intensity trend predicted by simulation for nanohole arrays with different nanohole periods. Moreover, the number of observed EVs in the best-performing nanohole array increases by more than 12 times compared with EVs immobilized on a reference substrate, uncovering a vast amount of weakly fluorescent EVs that would remain undetected with the regular fluorescent method. Our nanohole array provides a basis for a future platform of single-EV analyses, also promising to capture the signature arising from low-expressing proteins.
Collapse
Affiliation(s)
- Yupeng Yang
- Division
of Solid-State Electronics, Department of Electrical Engineering,
The Ångström Laboratory, Uppsala
University, SE-751 03 Uppsala, Sweden
| | - Prattakorn Metem
- Division
of Applied Electrochemistry, Department of Chemical Engineering, KTH Royal Institute of Technology, SE-100 44 Stockholm, Sweden
| | - Mohammad Hadi Khaksaran
- Division
of Solid-State Electronics, Department of Electrical Engineering,
The Ångström Laboratory, Uppsala
University, SE-751 03 Uppsala, Sweden
| | - Siddharth Sourabh Sahu
- Division
of Solid-State Electronics, Department of Electrical Engineering,
The Ångström Laboratory, Uppsala
University, SE-751 03 Uppsala, Sweden
| | - Fredrik Stridfeldt
- Bio-Opto-Nano
Physics, Department of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, SE-100 44 Stockholm, Sweden
| | - André Görgens
- Department
of Laboratory Medicine, Division of Biomolecular and Cellular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department
of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge and Karolinska
Comprehensive Cancer Center, 113 51 Stockholm, Sweden
- Institute
for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Shi-Li Zhang
- Division
of Solid-State Electronics, Department of Electrical Engineering,
The Ångström Laboratory, Uppsala
University, SE-751 03 Uppsala, Sweden
| | - Apurba Dev
- Division
of Solid-State Electronics, Department of Electrical Engineering,
The Ångström Laboratory, Uppsala
University, SE-751 03 Uppsala, Sweden
- Bio-Opto-Nano
Physics, Department of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, SE-100 44 Stockholm, Sweden
| |
Collapse
|
3
|
Sun S, Cox‐Vázquez SJ, Cho N, Bazan GC, Groves JT. Direct imaging with multidimensional labelling and high-content analysis allows quantitative categorization and characterizations of individual small extracellular vesicles and nanoparticles (sEVPs). J Extracell Vesicles 2024; 13:e12520. [PMID: 39665317 PMCID: PMC11635478 DOI: 10.1002/jev2.12520] [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: 11/02/2023] [Revised: 08/08/2024] [Accepted: 09/10/2024] [Indexed: 12/13/2024] Open
Abstract
Small extracellular vesicles and nanoparticles (sEVPs) are cell-secreted entities with potential as diagnostic biomarkers and therapeutic vehicles. However, significant intrinsic sEVP heterogeneity impedes analysis and understanding of their composition and functions. We employ multidimensional fluorescent labelling on sEVPs, leveraging the robustness of a newly developed membrane probe-conjugated oligoelectrolytes (COEs), and conduct total internal reflection fluorescence (TIRF) microscopy on sEVP arrays. These arrays comprise single sEVPs anchored to a soft material functionalized surface with little bias. We then develop an enhanced algorithm for colocalization analysis of the multiple labels on individual sEVPs and perform deep profiling of particle content. We categorize sEVPs derived from the same cell type into seven distinct subpopulations-some vesicular whereas others non-vesicular, and we demonstrate that sEVPs from four cell types exhibit quantitatively distinguishable subpopulation distributions. Furthermore, we gain insights into specific particle features within each subpopulation, including CD63 counts, relative particle size, relative concentration of cargoes, and correlations among different cargoes. This high-content analysis reveals common cargo sorting features in sEVP subpopulations across different cell types and suggests new statistics within the sEVP inherent heterogeneity that could differentiate sEVPs from two types of cancer cells and two types of normal cells. Collectively, our study presents a robust single-sEVP characterization platform, combining high-content imaging with comprehensive analysis. This platform is poised to advance sEVP-based theranostic assays and facilitate exploration into disease-associated sEVP biogenesis and sEVP-mediated intercellular communication.
Collapse
Affiliation(s)
- Simou Sun
- Institute for Digital Molecular Analytics and ScienceNanyang Technological UniversitySingaporeSingapore
- Current address: Department of ChemistryStony Brook UniversityNew YorkUnited States
| | - Sarah J. Cox‐Vázquez
- Department of ChemistryNational University of SingaporeSingaporeSingapore
- Institute for Functional Intelligent MaterialsNational University of SingaporeSingaporeSingapore
| | - Nam‐Joon Cho
- School of Materials Science and EngineeringNanyang Technological UniversitySingaporeSingapore
| | - Guillermo C. Bazan
- Institute for Digital Molecular Analytics and ScienceNanyang Technological UniversitySingaporeSingapore
- Department of ChemistryNational University of SingaporeSingaporeSingapore
- Institute for Functional Intelligent MaterialsNational University of SingaporeSingaporeSingapore
| | - Jay T. Groves
- Institute for Digital Molecular Analytics and ScienceNanyang Technological UniversitySingaporeSingapore
- Department of ChemistryUniversity of CaliforniaBerkeleyCaliforniaUSA
| |
Collapse
|
4
|
Walker SN, Lucas K, Dewey MJ, Badylak SF, Hussey GS, Flax J, McGrath JL. Rapid Assessment of Biomarkers on Single Extracellular Vesicles Using "Catch and Display" on Ultrathin Nanoporous Silicon Nitride Membranes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2405505. [PMID: 39358943 PMCID: PMC11961765 DOI: 10.1002/smll.202405505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/27/2024] [Indexed: 10/04/2024]
Abstract
Extracellular vesicles (EVs) are particles released from cells that facilitate intercellular communication and have tremendous diagnostic and therapeutic potential. Bulk assays lack the sensitivity to detect rare EV subsets relevant to disease, and while single EV analysis techniques remedy this, they are often undermined by complicated detection schemes and prohibitive instrumentation. To address these issues, a microfluidic technique for EV characterization called "catch and display for liquid biopsy (CAD-LB)" is proposed. In this method, minimally processed samples are pipette-injected and fluorescently labeled EVs are captured in the nanopores of an ultrathin membrane. This enables the rapid assessment of EV number and biomarker colocalization by light microscopy. Here, nanoparticles are used to define the accuracy and dynamic range for counting and colocalization. The same assessments are then made for purified EVs and for unpurified EVs in plasma. Biomarker detection is validated using CD9 and Western blot analysis to confirm that CAD-LB accurately reports relative protein expression levels. Using unprocessed conditioned media, CAD-LB captures the known increase in EV-associated ICAM-1 following endothelial cell cytokine stimulation. Finally, to demonstrate CAD-LB's clinical potential, EV biomarkers indicative of immunotherapy responsiveness are successfully detected in the plasma of bladder cancer patients treated with immune checkpoint blockade.
Collapse
Affiliation(s)
- Samuel N. Walker
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, United States
| | - Kilean Lucas
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, United States
| | - Marley J. Dewey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - George S. Hussey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - Jonathan Flax
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, United States, Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, United States
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, United States
| |
Collapse
|
5
|
Zhuang T, Wang S, Yu X, He X, Guo H, Ou C. Current status and future perspectives of platelet-derived extracellular vesicles in cancer diagnosis and treatment. Biomark Res 2024; 12:88. [PMID: 39183323 PMCID: PMC11346179 DOI: 10.1186/s40364-024-00639-0] [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: 06/29/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024] Open
Abstract
Platelets are a significant component of the cell population in the tumour microenvironment (TME). Platelets influence other immune cells and perform cross-talk with tumour cells, playing an important role in tumour development. Extracellular vesicles (EVs) are small membrane vesicles released from the cells into the TME. They can transfer biological information, including proteins, nucleic acids, and metabolites, from secretory cells to target receptor cells. This process affects the progression of various human diseases, particularly cancer. In recent years, several studies have demonstrated that platelet-derived extracellular vesicles (PEVs) can help regulate the malignant biological behaviours of tumours, including malignant proliferation, resistance to cell death, invasion and metastasis, metabolic reprogramming, immunity, and angiogenesis. Consequently, PEVs have been identified as key regulators of tumour progression. Therefore, targeting PEVs is a potential strategy for tumour treatment. Furthermore, the extensive use of nanomaterials in medical research has indicated that engineered PEVs are ideal delivery systems for therapeutic drugs. Recent studies have demonstrated that PEV engineering technologies play a pivotal role in the treatment of tumours by combining photothermal therapy, immunotherapy, and chemotherapy. In addition, aberrant changes in PEVs are closely associated with the clinicopathological features of patients with tumours, which may serve as liquid biopsy markers for early diagnosis, monitoring disease progression, and the prognostic assessment of patients with tumours. A comprehensive investigation into the role and potential mechanisms of PEVs in tumourigenesis may provide novel diagnostic biomarkers and potential therapeutic strategies for treating human tumours.
Collapse
Affiliation(s)
- Tongtao Zhuang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Shenrong Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaoqian Yu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaoyun He
- Departments of Ultrasound Imaging, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Hongbin Guo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Chunlin Ou
- 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.
| |
Collapse
|
6
|
Talebian Gevari M, Sahu SS, Stridfeldt F, Hååg P, De Petris L, Viktorsson K, Lewensohn R, Gori A, Cretich M, Dev A. Design and Optimization of a Silicon-Based Electrokinetic Microchip for Sensitive Detection of Small Extracellular Vesicles. ACS Sens 2024; 9:2935-2945. [PMID: 38848141 PMCID: PMC11217933 DOI: 10.1021/acssensors.4c00110] [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: 01/16/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024]
Abstract
Detection of analytes using streaming current has previously been explored using both experimental approaches and theoretical analyses of such data. However, further developments are needed for establishing a viable microchip that can be exploited to deliver a sensitive, robust, and scalable biosensor device. In this study, we demonstrated the fabrication of such a device on silicon wafer using a scalable silicon microfabrication technology followed by characterization and optimization of this sensor for detection of small extracellular vesicles (sEVs) with sizes in the range of 30 to 200 nm, as determined by nanoparticle tracking analyses. We showed that the sensitivity of the devices, assessed by a common protein-ligand pair and sEVs, significantly outperforms previous approaches using the same principle. Two versions of the microchips, denoted as enclosed and removable-top microchips, were developed and compared, aiming to discern the importance of high-pressure measurement versus easier and better surface preparation capacity. A custom-built chip manifold allowing easy interfacing with standard microfluidic connections was also constructed. By investigating different electrical, fluidic, morphological, and fluorescence measurements, we show that while the enclosed microchip with its robust glass-silicon bonding can withstand higher pressure and thus generate higher streaming current, the removable-top configuration offers several practical benefits, including easy surface preparation, uniform probe conjugation, and improvement in the limit of detection (LoD). We further compared two common surface functionalization strategies and showed that the developed microchip can achieve both high sensitivity for membrane protein profiling and low LoD for detection of sEV detection. At the optimum working condition, we demonstrated that the microchip could detect sEVs reaching an LoD of 104 sEVs/mL (when captured by membrane-sensing peptide (MSP) probes), which is among the lowest in the so far reported microchip-based methods.
Collapse
Affiliation(s)
- Moein Talebian Gevari
- Division
of Solid-State Electronics, Department of Electrical Engineering, Uppsala University, 75 121 Uppsala, Sweden
| | - Siddharth Sourabh Sahu
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, 10 691 Stockholm, Sweden
| | - Fredrik Stridfeldt
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, 10 691 Stockholm, Sweden
| | - Petra Hååg
- Department
of Oncology-Pathology, Karolinska Institutet, 171 64 Solna, Sweden
| | - Luigi De Petris
- Department
of Oncology-Pathology, Karolinska Institutet, 171 64 Solna, Sweden
- Theme
Cancer, Medical Unit Head and Neck, Lung, and Skin Tumors, Thoracic
Oncology Center, Karolinska University Hospital, 171 64 Solna, Sweden
| | - Kristina Viktorsson
- Department
of Oncology-Pathology, Karolinska Institutet, 171 64 Solna, Sweden
| | - Rolf Lewensohn
- Department
of Oncology-Pathology, Karolinska Institutet, 171 64 Solna, Sweden
- Theme
Cancer, Medical Unit Head and Neck, Lung, and Skin Tumors, Thoracic
Oncology Center, Karolinska University Hospital, 171 64 Solna, Sweden
| | - Alessandro Gori
- Consiglio
Nazionale delle Ricerche, Istituto di Scienze
e Tecnologie Chimiche “Giulio Natta” (SCITEC), 20131 Milan, Italy
| | - Marina Cretich
- Consiglio
Nazionale delle Ricerche, Istituto di Scienze
e Tecnologie Chimiche “Giulio Natta” (SCITEC), 20131 Milan, Italy
| | - Apurba Dev
- Division
of Solid-State Electronics, Department of Electrical Engineering, Uppsala University, 75 121 Uppsala, Sweden
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, 10 691 Stockholm, Sweden
| |
Collapse
|
7
|
Nguyen VVT, Welsh JA, Tertel T, Choo A, van de Wakker SI, Defourny KAY, Giebel B, Vader P, Padmanabhan J, Lim SK, Nolte‐'t Hoen ENM, Verhaar MC, Bostancioglu RB, Zickler AM, Hong JM, Jones JC, EL Andaloussi S, van Balkom BWM, Görgens A. Inter-laboratory multiplex bead-based surface protein profiling of MSC-derived EV preparations identifies MSC-EV surface marker signatures. J Extracell Vesicles 2024; 13:e12463. [PMID: 38868945 PMCID: PMC11170075 DOI: 10.1002/jev2.12463] [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: 10/20/2023] [Revised: 04/15/2024] [Accepted: 05/21/2024] [Indexed: 06/14/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) are promising regenerative therapeutics that primarily exert their effects through secreted extracellular vesicles (EVs). These EVs - being small and non-living - are easier to handle and possess advantages over cellular products. Consequently, the therapeutic potential of MSC-EVs is increasingly investigated. However, due to variations in MSC-EV manufacturing strategies, MSC-EV products should be considered as highly diverse. Moreover, the diverse array of EV characterisation technologies used for MSC-EV characterisation further complicates reliable interlaboratory comparisons of published data. Consequently, this study aimed to establish a common method that can easily be used by various MSC-EV researchers to characterise MSC-EV preparations to facilitate interlaboratory comparisons. To this end, we conducted a comprehensive inter-laboratory assessment using a novel multiplex bead-based EV flow cytometry assay panel. This assessment involved 11 different MSC-EV products from five laboratories with varying MSC sources, culture conditions, and EV preparation methods. Through this assay panel covering a range of mostly MSC-related markers, we identified a set of cell surface markers consistently positive (CD44, CD73 and CD105) or negative (CD11b, CD45 and CD197) on EVs of all explored MSC-EV preparations. Hierarchical clustering analysis revealed distinct surface marker profiles associated with specific preparation processes and laboratory conditions. We propose CD73, CD105 and CD44 as robust positive markers for minimally identifying MSC-derived EVs and CD11b, CD14, CD19, CD45 and CD79 as reliable negative markers. Additionally, we highlight the influence of culture medium components, particularly human platelet lysate, on EV surface marker profiles, underscoring the influence of culture conditions on resulting EV products. This standardisable approach for MSC-EV surface marker profiling offers a tool for routine characterisation of manufactured EV products in pre-clinical and clinical research, enhances the quality control of MSC-EV preparations, and hopefully paves the way for higher consistency and reproducibility in the emerging therapeutic MSC-EV field.
Collapse
Affiliation(s)
| | - Joshua A. Welsh
- Translational Nanobiology Section, Laboratory of Pathology, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
- The Measuring Stick, LtdPeterboroughUK
- Advanced Technology GroupBecton DickinsonSan JoseCaliforniaUSA
| | - Tobias Tertel
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Andre Choo
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Simonides I. van de Wakker
- Department of Cardiology, Experimental Cardiology LaboratoryUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Kyra A. Y. Defourny
- Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Bernd Giebel
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Pieter Vader
- Department of Cardiology, Experimental Cardiology LaboratoryUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
- CDL ResearchUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Jayanthi Padmanabhan
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Sai Kiang Lim
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Esther N. M. Nolte‐'t Hoen
- Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | | | - R. Beklem Bostancioglu
- Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
| | - Antje M. Zickler
- Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
- Karolinska ATMP CenterANA FuturaHuddingeSweden
| | - Jia Mei Hong
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Jennifer C. Jones
- Translational Nanobiology Section, Laboratory of Pathology, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Samir EL Andaloussi
- Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
- Karolinska ATMP CenterANA FuturaHuddingeSweden
| | | | - André Görgens
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
- Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
- Karolinska ATMP CenterANA FuturaHuddingeSweden
| |
Collapse
|
8
|
Walker SN, Lucas K, Dewey MJ, Badylak S, Hussey G, Flax J, McGrath JL. Rapid Assessment of Biomarkers on Single Extracellular Vesicles Using 'Catch and Display' on Ultrathin Nanoporous Silicon Nitride Membranes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.29.589900. [PMID: 38746341 PMCID: PMC11092443 DOI: 10.1101/2024.04.29.589900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Extracellular vesicles (EVs) are particles secreted by all cells that carry bioactive cargo and facilitate intercellular communication with roles in normal physiology and disease pathogenesis. EVs have tremendous diagnostic and therapeutic potential and accordingly, the EV field has grown exponentially in recent years. Bulk assays lack the sensitivity to detect rare EV subsets relevant to disease, and while single EV analysis techniques remedy this, they are undermined by complicated detection schemes often coupled with prohibitive instrumentation. To address these issues, we propose a microfluidic technique for EV characterization called 'catch and display for liquid biopsy (CAD-LB)'. CAD-LB rapidly captures fluorescently labeled EVs in the similarly-sized pores of an ultrathin silicon nitride membrane. Minimally processed sample is introduced via pipette injection into a simple microfluidic device which is directly imaged using fluorescence microscopy for a rapid assessment of EV number and biomarker colocalization. In this work, nanoparticles were first used to define the accuracy and dynamic range for counting and colocalization by CAD-LB. Following this, the same assessments were made for purified EVs and for unpurified EVs in plasma. Biomarker detection was validated using CD9 in which Western blot analysis confirmed that CAD-LB faithfully recapitulated differing expression levels among samples. We further verified that CAD-LB captured the known increase in EV-associated ICAM-1 following the cytokine stimulation of endothelial cells. Finally, to demonstrate CAD-LB's clinical potential, we show that EV biomarkers indicative of immunotherapy responsiveness are successfully detected in the plasma of bladder cancer patients undergoing immune checkpoint blockade.
Collapse
Affiliation(s)
- Samuel N. Walker
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, United States
| | - Kilean Lucas
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, United States
| | - Marley J. Dewey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - Stephen Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - George Hussey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - Jonathan Flax
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, United States
- Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, United States
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, United States
| |
Collapse
|
9
|
Vo N, Tran C, Tran NHB, Nguyen NT, Nguyen T, Ho DTK, Nguyen DDN, Pham T, Nguyen TA, Phan HTN, Nguyen H, Tu LN. A novel multi-stage enrichment workflow and comprehensive characterization for HEK293F-derived extracellular vesicles. J Extracell Vesicles 2024; 13:e12454. [PMID: 38760878 PMCID: PMC11101607 DOI: 10.1002/jev2.12454] [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: 10/19/2023] [Revised: 03/14/2024] [Accepted: 05/03/2024] [Indexed: 05/19/2024] Open
Abstract
Extracellular vesicles (EVs) are emerging as a promising drug delivery vehicle as they are biocompatible and capable of targeted delivery. However, clinical translation of EVs remains challenging due to the lack of standardized and scalable manufacturing protocols to consistently isolate small EVs (sEVs) with both high yield and high purity. The heterogenous nature of sEVs leading to unknown composition of biocargos causes further pushback due to safety concerns. In order to address these issues, we developed a robust quality-controlled multi-stage process to produce and isolate sEVs from human embryonic kidney HEK293F cells. We then compared different 2-step and 3-step workflows for eliminating protein impurities and cell-free nucleic acids to meet acceptable limits of regulatory authorities. Our results showed that sEV production was maximized when HEK293F cells were grown at high-density stationary phase in semi-continuous culture. The novel 3-step workflow combining tangential flow filtration, sucrose-cushion ultracentrifugation and bind-elute size-exclusion chromatography outperformed other methods in sEV purity while still preserved high yield and particle integrity. The purified HEK293F-derived sEVs were thoroughly characterized for identity including sub-population analysis, content profiling including proteomics and miRNA sequencing, and demonstrated excellent preclinical safety profile in both in-vitro and in-vivo testing. Our rigorous enrichment workflow and comprehensive characterization will help advance the development of EVs, particularly HEK293F-derived sEVs, to be safe and reliable drug carriers for therapeutic applications.
Collapse
Affiliation(s)
- Nhan Vo
- Medical Genetics InstituteHo Chi Minh CityVietnam
| | - Chau Tran
- Medical Genetics InstituteHo Chi Minh CityVietnam
| | | | | | - Thieu Nguyen
- Medical Genetics InstituteHo Chi Minh CityVietnam
| | | | | | - Tran Pham
- Medical Genetics InstituteHo Chi Minh CityVietnam
| | | | | | | | - Lan N. Tu
- Medical Genetics InstituteHo Chi Minh CityVietnam
| |
Collapse
|
10
|
Das S, Lyon CJ, Hu T. A Panorama of Extracellular Vesicle Applications: From Biomarker Detection to Therapeutics. ACS NANO 2024; 18:9784-9797. [PMID: 38471757 PMCID: PMC11008359 DOI: 10.1021/acsnano.4c00666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/04/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024]
Abstract
Extracellular vesicles (EVs) secreted by all cell types are involved in the cell-to-cell transfer of regulatory factors that influence cell and tissue phenotypes in normal and diseased tissues. EVs are thus a rich source of biomarker targets for assays that analyze blood and urinary EVs for disease diagnosis. Sensitive biomarker detection in EVs derived from specific cell populations is a key major hurdle when analyzing complex biological samples, but innovative approaches surveyed in this Perspective can streamline EV isolation and enhance the sensitivity of EV detection procedures required for clinical application of EV-based diagnostics and therapeutics, including nanotechnology and microfluidics, to achieve EV characterizations. Finally, this Perspective also outlines opportunities and challenges remaining for clinical translation of EV-based assays.
Collapse
Affiliation(s)
- Sumita Das
- Center for Cellular and Molecular Diagnostics
and Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Christopher J. Lyon
- Center for Cellular and Molecular Diagnostics
and Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Tony Hu
- Center for Cellular and Molecular Diagnostics
and Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| |
Collapse
|
11
|
Zhang XW, Qi GX, Liu MX, Yang YF, Wang JH, Yu YL, Chen S. Deep Learning Promotes Profiling of Multiple miRNAs in Single Extracellular Vesicles for Cancer Diagnosis. ACS Sens 2024; 9:1555-1564. [PMID: 38442411 DOI: 10.1021/acssensors.3c02789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Extracellular vesicle microRNAs (EV miRNAs) are critical noninvasive biomarkers for early cancer diagnosis. However, accurate cancer diagnosis based on bulk analysis is hindered by the heterogeneity among EVs. Herein, we report an approach for profiling single-EV multi-miRNA signatures by combining total internal reflection fluorescence (TIRF) imaging with a deep learning (DL) algorithm for the first time. This innovative technique allows for the precise characterization of EV miRNAs at the single-vesicle level, overcoming the challenges posed by EV heterogeneity. TIRF with high resolution and a signal-to-noise ratio can simultaneously detect multi-miRNAs in situ in individual EVs. DL algorithm avoids complicated and inaccurate artificial feature extraction, achieving automated high-resolution image analysis. Using this approach, we reveal that the main variation of EVs from 5 cancer cells and normal plasma is the triple-positive EV subpopulation, and the classification accuracy of single triple-positive EVs from 6 sources can reach above 95%. In the clinical cohort, 20 patients (5 lung cancer, 5 breast cancer, 5 cervical cancer, and 5 colon cancer) and 5 healthy controls are predicted with an overall accuracy of 100%. This single-EV strategy provides new opportunities for exploring more specific EV biomarkers to achieve cancer diagnosis and classification.
Collapse
Affiliation(s)
- Xue-Wei Zhang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Gong-Xiang Qi
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Meng-Xian Liu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Yan-Fei Yang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Jian-Hua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Yong-Liang Yu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Shuai Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| |
Collapse
|
12
|
Welsh JA, Goberdhan DCI, O'Driscoll L, Buzas EI, Blenkiron C, Bussolati B, Cai H, Di Vizio D, Driedonks TAP, Erdbrügger U, Falcon‐Perez JM, Fu Q, Hill AF, Lenassi M, Lim SK, Mahoney MG, Mohanty S, Möller A, Nieuwland R, Ochiya T, Sahoo S, Torrecilhas AC, Zheng L, Zijlstra A, Abuelreich S, Bagabas R, Bergese P, Bridges EM, Brucale M, Burger D, Carney RP, Cocucci E, Colombo F, Crescitelli R, Hanser E, Harris AL, Haughey NJ, Hendrix A, Ivanov AR, Jovanovic‐Talisman T, Kruh‐Garcia NA, Ku'ulei‐Lyn Faustino V, Kyburz D, Lässer C, Lennon KM, Lötvall J, Maddox AL, Martens‐Uzunova ES, Mizenko RR, Newman LA, Ridolfi A, Rohde E, Rojalin T, Rowland A, Saftics A, Sandau US, Saugstad JA, Shekari F, Swift S, Ter‐Ovanesyan D, Tosar JP, Useckaite Z, Valle F, Varga Z, van der Pol E, van Herwijnen MJC, Wauben MHM, Wehman AM, Williams S, Zendrini A, Zimmerman AJ, MISEV Consortium, Théry C, Witwer KW. Minimal information for studies of extracellular vesicles (MISEV2023): From basic to advanced approaches. J Extracell Vesicles 2024; 13:e12404. [PMID: 38326288 PMCID: PMC10850029 DOI: 10.1002/jev2.12404] [Citation(s) in RCA: 1038] [Impact Index Per Article: 1038.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 02/09/2024] Open
Abstract
Extracellular vesicles (EVs), through their complex cargo, can reflect the state of their cell of origin and change the functions and phenotypes of other cells. These features indicate strong biomarker and therapeutic potential and have generated broad interest, as evidenced by the steady year-on-year increase in the numbers of scientific publications about EVs. Important advances have been made in EV metrology and in understanding and applying EV biology. However, hurdles remain to realising the potential of EVs in domains ranging from basic biology to clinical applications due to challenges in EV nomenclature, separation from non-vesicular extracellular particles, characterisation and functional studies. To address the challenges and opportunities in this rapidly evolving field, the International Society for Extracellular Vesicles (ISEV) updates its 'Minimal Information for Studies of Extracellular Vesicles', which was first published in 2014 and then in 2018 as MISEV2014 and MISEV2018, respectively. The goal of the current document, MISEV2023, is to provide researchers with an updated snapshot of available approaches and their advantages and limitations for production, separation and characterisation of EVs from multiple sources, including cell culture, body fluids and solid tissues. In addition to presenting the latest state of the art in basic principles of EV research, this document also covers advanced techniques and approaches that are currently expanding the boundaries of the field. MISEV2023 also includes new sections on EV release and uptake and a brief discussion of in vivo approaches to study EVs. Compiling feedback from ISEV expert task forces and more than 1000 researchers, this document conveys the current state of EV research to facilitate robust scientific discoveries and move the field forward even more rapidly.
Collapse
Affiliation(s)
- Joshua A. Welsh
- Translational Nanobiology Section, Laboratory of PathologyNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Deborah C. I. Goberdhan
- Nuffield Department of Women's and Reproductive HealthUniversity of Oxford, Women's Centre, John Radcliffe HospitalOxfordUK
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical SciencesTrinity College DublinDublinIreland
- Trinity Biomedical Sciences InstituteTrinity College DublinDublinIreland
- Trinity St. James's Cancer InstituteTrinity College DublinDublinIreland
| | - Edit I. Buzas
- Department of Genetics, Cell‐ and ImmunobiologySemmelweis UniversityBudapestHungary
- HCEMM‐SU Extracellular Vesicle Research GroupSemmelweis UniversityBudapestHungary
- HUN‐REN‐SU Translational Extracellular Vesicle Research GroupSemmelweis UniversityBudapestHungary
| | - Cherie Blenkiron
- Faculty of Medical and Health SciencesThe University of AucklandAucklandNew Zealand
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health SciencesUniversity of TurinTurinItaly
| | | | - Dolores Di Vizio
- Department of Surgery, Division of Cancer Biology and TherapeuticsCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Tom A. P. Driedonks
- Department CDL ResearchUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Uta Erdbrügger
- University of Virginia Health SystemCharlottesvilleVirginiaUSA
| | - Juan M. Falcon‐Perez
- Exosomes Laboratory, Center for Cooperative Research in BiosciencesBasque Research and Technology AllianceDerioSpain
- Metabolomics Platform, Center for Cooperative Research in BiosciencesBasque Research and Technology AllianceDerioSpain
- IKERBASQUE, Basque Foundation for ScienceBilbaoSpain
| | - Qing‐Ling Fu
- Otorhinolaryngology Hospital, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Extracellular Vesicle Research and Clinical Translational CenterThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Andrew F. Hill
- Institute for Health and SportVictoria UniversityMelbourneAustralia
| | - Metka Lenassi
- Faculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Sai Kiang Lim
- Institute of Molecular and Cell Biology (IMCB)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
- Paracrine Therapeutics Pte. Ltd.SingaporeSingapore
- Department of Surgery, YLL School of MedicineNational University SingaporeSingaporeSingapore
| | - Mỹ G. Mahoney
- Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Sujata Mohanty
- Stem Cell FacilityAll India Institute of Medical SciencesNew DelhiIndia
| | - Andreas Möller
- Chinese University of Hong KongHong KongHong Kong S.A.R.
- QIMR Berghofer Medical Research InstituteBrisbaneAustralia
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
| | | | - Susmita Sahoo
- Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ana C. Torrecilhas
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais, Químicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP) Campus DiademaDiademaBrazil
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Andries Zijlstra
- Department of PathologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- GenentechSouth San FranciscoCaliforniaUSA
| | - Sarah Abuelreich
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Reem Bagabas
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Paolo Bergese
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- Center for Colloid and Surface Science (CSGI)FlorenceItaly
- National Center for Gene Therapy and Drugs based on RNA TechnologyPaduaItaly
| | - Esther M. Bridges
- Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| | - Marco Brucale
- Consiglio Nazionale delle Ricerche ‐ Istituto per lo Studio dei Materiali NanostrutturatiBolognaItaly
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande InterfaseFlorenceItaly
| | - Dylan Burger
- Kidney Research CentreOttawa Hopsital Research InstituteOttawaCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaCanada
- School of Pharmaceutical SciencesUniversity of OttawaOttawaCanada
| | - Randy P. Carney
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Emanuele Cocucci
- Division of Pharmaceutics and Pharmacology, College of PharmacyThe Ohio State UniversityColumbusOhioUSA
- Comprehensive Cancer CenterThe Ohio State UniversityColumbusOhioUSA
| | - Federico Colombo
- Division of Pharmaceutics and Pharmacology, College of PharmacyThe Ohio State UniversityColumbusOhioUSA
| | - Rossella Crescitelli
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Edveena Hanser
- Department of BiomedicineUniversity Hospital BaselBaselSwitzerland
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | | | - Norman J. Haughey
- Departments of Neurology and PsychiatryJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Human Structure and RepairGhent UniversityGhentBelgium
- Cancer Research Institute GhentGhentBelgium
| | - Alexander R. Ivanov
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | - Tijana Jovanovic‐Talisman
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Nicole A. Kruh‐Garcia
- Bio‐pharmaceutical Manufacturing and Academic Resource Center (BioMARC)Infectious Disease Research Center, Colorado State UniversityFort CollinsColoradoUSA
| | - Vroniqa Ku'ulei‐Lyn Faustino
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Diego Kyburz
- Department of BiomedicineUniversity of BaselBaselSwitzerland
- Department of RheumatologyUniversity Hospital BaselBaselSwitzerland
| | - Cecilia Lässer
- Krefting Research Centre, Department of Internal Medicine and Clinical NutritionInstitute of Medicine at Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Kathleen M. Lennon
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Adam L. Maddox
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Elena S. Martens‐Uzunova
- Erasmus MC Cancer InstituteUniversity Medical Center Rotterdam, Department of UrologyRotterdamThe Netherlands
| | - Rachel R. Mizenko
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Lauren A. Newman
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Andrea Ridolfi
- Department of Physics and Astronomy, and LaserLaB AmsterdamVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Eva Rohde
- Department of Transfusion Medicine, University HospitalSalzburger Landeskliniken GmbH of Paracelsus Medical UniversitySalzburgAustria
- GMP Unit, Paracelsus Medical UniversitySalzburgAustria
- Transfer Centre for Extracellular Vesicle Theralytic Technologies, EV‐TTSalzburgAustria
| | - Tatu Rojalin
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
- Expansion Therapeutics, Structural Biology and BiophysicsJupiterFloridaUSA
| | - Andrew Rowland
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Andras Saftics
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Ursula S. Sandau
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Julie A. Saugstad
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Celer DiagnosticsTorontoCanada
| | - Simon Swift
- Waipapa Taumata Rau University of AucklandAucklandNew Zealand
| | - Dmitry Ter‐Ovanesyan
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMassachusettsUSA
| | - Juan P. Tosar
- Universidad de la RepúblicaMontevideoUruguay
- Institut Pasteur de MontevideoMontevideoUruguay
| | - Zivile Useckaite
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Francesco Valle
- Consiglio Nazionale delle Ricerche ‐ Istituto per lo Studio dei Materiali NanostrutturatiBolognaItaly
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande InterfaseFlorenceItaly
| | - Zoltan Varga
- Biological Nanochemistry Research GroupInstitute of Materials and Environmental Chemistry, Research Centre for Natural SciencesBudapestHungary
- Department of Biophysics and Radiation BiologySemmelweis UniversityBudapestHungary
| | - Edwin van der Pol
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Biomedical Engineering and Physics, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Martijn J. C. van Herwijnen
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Marca H. M. Wauben
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | | | | | - Andrea Zendrini
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- Center for Colloid and Surface Science (CSGI)FlorenceItaly
| | - Alan J. Zimmerman
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | | | - Clotilde Théry
- Institut Curie, INSERM U932PSL UniversityParisFrance
- CurieCoreTech Extracellular Vesicles, Institut CurieParisFrance
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- EV Core Facility “EXCEL”, Institute for Basic Biomedical SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
13
|
Jang HJ, Shim KS, Lee J, Park JH, Kang SJ, Shin YM, Lee JB, Baek W, Yoon JK. Engineering of Cell Derived-Nanovesicle as an Alternative to Exosome Therapy. Tissue Eng Regen Med 2024; 21:1-19. [PMID: 38066355 PMCID: PMC10764700 DOI: 10.1007/s13770-023-00610-4] [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: 10/04/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 01/04/2024] Open
Abstract
BACKGROUND Exosomes, nano-sized vesicles ranging between 30 and 150 nm secreted by human cells, play a pivotal role in long-range intercellular communication and have attracted significant attention in the field of regenerative medicine. Nevertheless, their limited productivity and cost-effectiveness pose challenges for clinical applications. These issues have recently been addressed by cell-derived nanovesicles (CDNs), which are physically synthesized exosome-mimetic nanovesicles from parent cells, as a promising alternative to exosomes. CDNs exhibit structural, physical, and biological properties similar to exosomes, containing intracellular protein and genetic components encapsulated by the cell plasma membrane. These characteristics allow CDNs to be used as regenerative medicine and therapeutics on their own, or as a drug delivery system. METHODS The paper reviews diverse methods for CDN synthesis, current analysis techniques, and presents engineering strategies to improve lesion targeting efficiency and/or therapeutic efficacy. RESULTS CDNs, with their properties similar to those of exosomes, offer a cost-effective and highly productive alternative due to their non-living biomaterial nature, nano-size, and readiness for use, allowing them to overcome several limitations of conventional cell therapy methods. CONCLUSION Ongoing research and enhancement of CDNs engineering, along with comprehensive safety assessments and stability analysis, exhibit vast potential to advance regenerative medicine by enabling the development of efficient therapeutic interventions.
Collapse
Affiliation(s)
- Hye-Jeong Jang
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea
| | - Kyu-Sik Shim
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jinah Lee
- Department of Biological Science, Research Institute of Women's Health, Brain Korea 21 Project, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Joo Hyeon Park
- Department of Biological Science, Research Institute of Women's Health, Brain Korea 21 Project, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Seong-Jun Kang
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea
| | - Young Min Shin
- Department of Biological Science, Research Institute of Women's Health, Brain Korea 21 Project, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Jung Bok Lee
- Department of Biological Science, Research Institute of Women's Health, Brain Korea 21 Project, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| | - Wooyeol Baek
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea.
| |
Collapse
|
14
|
Ortiz-Perez A, Zhang M, Fitzpatrick LW, Izquierdo-Lozano C, Albertazzi L. Advanced optical imaging for the rational design of nanomedicines. Adv Drug Deliv Rev 2024; 204:115138. [PMID: 37980951 DOI: 10.1016/j.addr.2023.115138] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/21/2023]
Abstract
Despite the enormous potential of nanomedicines to shape the future of medicine, their clinical translation remains suboptimal. Translational challenges are present in every step of the development pipeline, from a lack of understanding of patient heterogeneity to insufficient insights on nanoparticle properties and their impact on material-cell interactions. Here, we discuss how the adoption of advanced optical microscopy techniques, such as super-resolution optical microscopies, correlative techniques, and high-content modalities, could aid the rational design of nanocarriers, by characterizing the cell, the nanomaterial, and their interaction with unprecedented spatial and/or temporal detail. In this nanomedicine arena, we will discuss how the implementation of these techniques, with their versatility and specificity, can yield high volumes of multi-parametric data; and how machine learning can aid the rapid advances in microscopy: from image acquisition to data interpretation.
Collapse
Affiliation(s)
- Ana Ortiz-Perez
- Department of Biomedical Engineering, Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Miao Zhang
- Department of Biomedical Engineering, Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Laurence W Fitzpatrick
- Department of Biomedical Engineering, Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Cristina Izquierdo-Lozano
- Department of Biomedical Engineering, Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Lorenzo Albertazzi
- Department of Biomedical Engineering, Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
15
|
Lee S, Verkhoturov DS, Eller MJ, Verkhoturov SV, Shaw MA, Gwon K, Kim Y, Lucien F, Malhi H, Revzin A, Schweikert EA. Nanoprojectile Secondary Ion Mass Spectrometry Enables Multiplexed Analysis of Individual Hepatic Extracellular Vesicles. ACS NANO 2023; 17:23584-23594. [PMID: 38033295 PMCID: PMC10985841 DOI: 10.1021/acsnano.3c06604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Extracellular vesicles (EVs) are nanoscale lipid bilayer particles secreted by cells. EVs may carry markers of the tissue of origin and its disease state, which makes them incredibly promising for disease diagnosis and surveillance. While the armamentarium of EV analysis technologies is rapidly expanding, there remains a strong need for multiparametric analysis with single EV resolution. Nanoprojectile (NP) secondary ion mass spectrometry (NP-SIMS) relies on bombarding a substrate of interest with individual gold NPs resolved in time and space. Each projectile creates an impact crater of 10-20 nm in diameter while molecules emitted from each impact are mass analyzed and recorded as individual mass spectra. We demonstrate the utility of NP-SIMS for statistical analysis of single EVs derived from normal liver cells (hepatocytes) and liver cancer cells. EVs were captured on antibody (Ab)-functionalized gold substrate and then labeled with Abs carrying lanthanide (Ln) MS tags (Ab@Ln). These tags targeted four markers selected for identifying all EVs, and specific to hepatocytes or liver cancer. NP-SIMS was used to detect Ab@Ln-tags colocalized on the same EV and to construct scatter plots of surface marker expression for thousands of EVs with the capability of categorizing individual EVs. Additionally, NP-SIMS revealed information about the chemical nanoenvironment where targeted moieties colocalized. Our approach allowed analysis of population heterogeneity with single EV resolution and distinguishing between hepatocyte and liver cancer EVs based on surface marker expression. NP-SIMS holds considerable promise for multiplexed analysis of single EVs and may become a valuable tool for identifying and validating EV biomarkers of cancer and other diseases.
Collapse
Affiliation(s)
- Seonhwa Lee
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Michael J. Eller
- Department of Chemistry and Biochemistry, California State University Northridge, Northridge, CA 91330, USA
| | | | - Michael A. Shaw
- Department of Chemistry and Biochemistry, California State University Northridge, Northridge, CA 91330, USA
| | - Kihak Gwon
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yohan Kim
- Departments of Urology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Fabrice Lucien
- Departments of Urology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Emile A. Schweikert
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, USA
| |
Collapse
|
16
|
Reynolds DE, Pan M, Yang J, Galanis G, Roh YH, Morales RT, Kumar SS, Heo S, Xu X, Guo W, Ko J. Double Digital Assay for Single Extracellular Vesicle and Single Molecule Detection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303619. [PMID: 37802976 PMCID: PMC10667851 DOI: 10.1002/advs.202303619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 09/13/2023] [Indexed: 10/08/2023]
Abstract
Extracellular vesicles (EVs) have emerged as a promising source of biomarkers for disease diagnosis. However, current diagnostic methods for EVs present formidable challenges, given the low expression levels of biomarkers carried by EV samples, as well as their complex physical and biological properties. Herein, a highly sensitive double digital assay is developed that allows for the absolute quantification of individual molecules from a single EV. Because the relative abundance of proteins is low for a single EV, tyramide signal amplification (TSA) is integrated to increase the fluorescent signal readout for evaluation. With the integrative microfluidic technology, the technology's ability to compartmentalize single EVs is successfully demonstrated, proving the technology's digital partitioning capacity. Then the device is applied to detect single PD-L1 proteins from single EVs derived from a melanoma cell line and it is discovered that there are ≈2.7 molecules expressed per EV, demonstrating the applicability of the system for profiling important prognostic and diagnostic cancer biomarkers for therapy response, metastatic status, and tumor progression. The ability to accurately quantify protein molecules of rare abundance from individual EVs will shed light on the understanding of EV heterogeneity and discovery of EV subtypes as new biomarkers.
Collapse
Affiliation(s)
- David E. Reynolds
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Menghan Pan
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Jingbo Yang
- Department of Pathology and Laboratory MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - George Galanis
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Yoon Ho Roh
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | | | | | - Su‐Jin Heo
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Department of Orthopaedic SurgeryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Xiaowei Xu
- Department of Pathology and Laboratory MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Wei Guo
- Department of BiologySchool of Arts and SciencesUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Jina Ko
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Department of Pathology and Laboratory MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| |
Collapse
|
17
|
Agrawal V, Pandey V, Mitra D. Active buckling of pressurized spherical shells: Monte Carlo simulation. Phys Rev E 2023; 108:L032601. [PMID: 37849090 DOI: 10.1103/physreve.108.l032601] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/06/2023] [Indexed: 10/19/2023]
Abstract
We study the buckling of pressurized spherical shells by Monte Carlo simulations in which the detailed balance is explicitly broken-thereby driving the shell to be active, out of thermal equilibrium. Such a shell typically has either higher (active) or lower (sedate) fluctuations compared to one in thermal equilibrium depending on how the detailed balance is broken. We show that, for the same set of elastic parameters, a shell that is not buckled in thermal equilibrium can be buckled if turned active. Similarly a shell that is buckled in thermal equilibrium can unbuckle if sedated. Based on this result, we suggest that it is possible to experimentally design microscopic elastic shells whose buckling can be optically controlled.
Collapse
Affiliation(s)
- Vipin Agrawal
- Nordita, KTH Royal Institute of Technology and Stockholm University, Hannes Alfvéns väg 12, 106 91 Stockholm, Sweden
- Department of Physics, Stockholm University, AlbaNova University Centre, Fysikum, 106 91 Stockholm, Sweden
| | - Vikash Pandey
- Nordita, KTH Royal Institute of Technology and Stockholm University, Hannes Alfvéns väg 12, 106 91 Stockholm, Sweden
| | - Dhrubaditya Mitra
- Nordita, KTH Royal Institute of Technology and Stockholm University, Hannes Alfvéns väg 12, 106 91 Stockholm, Sweden
| |
Collapse
|
18
|
Wang J, Cong L, Shi W, Xu W, Xu S. Single-Cell Analysis and Classification according to Multiplexed Proteins via Microdroplet-Based Self-Driven Magnetic Surface-Enhanced Raman Spectroscopy Platforms Assisted with Machine Learning Algorithms. Anal Chem 2023. [PMID: 37419505 DOI: 10.1021/acs.analchem.3c01273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2023]
Abstract
A microdroplet-based surface-enhanced Raman spectroscopy (microdroplet SERS) platform was constructed to envelop individual cells in microdroplets, followed by the SERS detection of their extracellular vesicle-proteins (EV-proteins) via the in-drop immunoassays by use of immunomagnetic beads (iMBs) and immuno-SERS tags (iSERS tags). A unique phenomenon is found that iMBs can start a spontaneous reorientation on the probed cell surface based on the electrostatic force-driven interfacial aggregation effect, which leads EV-proteins and iSERS tags to be gathered from a liquid phase to a cell membrane interface and significantly improves SERS sensitivity to the single-cell analysis level due to the formation of numbers of SERS hotspots. Three EV-proteins from two breast cancer cell lines were collected and further analyzed by machine learning algorithmic tools, which will be helpful for a deeper understanding of breast cancer subtypes from the view of EV-proteins.
Collapse
Affiliation(s)
- Jiaqi Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Lili Cong
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Wei Shi
- Key Lab for Molecular Enzymology & Engineering of Ministry of Education, Jilin University, Changchun 130012, P. R. China
| | - Weiqing Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Institute of Theoretical Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Shuping Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- State Key Laboratory of Applied Optics, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun 130033, P. R. China
- Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Institute of Theoretical Chemistry, Jilin University, Changchun 130012, P. R. China
| |
Collapse
|
19
|
Sahu SS, Gevari MT, Nagy Á, Gestin M, Hååg P, Lewensohn R, Viktorsson K, Karlström AE, Dev A. Multi-marker profiling of extracellular vesicles using streaming current and sequential electrostatic labeling. Biosens Bioelectron 2023; 227:115142. [PMID: 36805937 DOI: 10.1016/j.bios.2023.115142] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/24/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
High heterogeneity in the membrane protein expression of small extracellular vesicles (sEVs) means that bulk methods relying on antibody-based capture for expression analysis have a drawback that each type of antibody may capture a different sub-population. An improved approach is to capture a representative sEV population, without any bias, and then perform a multiplexed protein expression analysis on this population. However, such a possibility has been largely limited to fluorescence-based methods. Here, we present a novel electrostatic labelling strategy and a microchip-based all-electric method for membrane protein analysis of sEVs. The method allows us to profile multiple surface proteins on the captured sEVs using alternating charge labels. It also permits the comparison of expression levels in different sEV-subtypes. The proof of concept was tested by capturing sEVs both non-specifically (unbiased) as well as via anti-CD9 capture probes (biased), and then profiling the expression levels of various surface proteins using the charge labelled antibodies. The method is the first of its kind, demonstrating an all-electrical and microchip based method that allows for unbiased analysis of sEV membrane protein expression, comparison of expression levels in different sEV subsets, and fractional estimation of different sEV sub-populations. These results were also validated in parallel using a single-sEV fluorescence technique.
Collapse
Affiliation(s)
- Siddharth S Sahu
- Department of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, 10691, Stockholm, Sweden.
| | - Moein T Gevari
- Division of Solid-State Electronics, Department of Electrical Engineering, Uppsala University, 75121, Uppsala, Sweden
| | - Ábel Nagy
- Department of Protein Science, School of Chemistry, Biotechnology, and Health (CBH), KTH Royal Institute of Technology, Stockholm, Sweden
| | - Maxime Gestin
- Department of Protein Science, School of Chemistry, Biotechnology, and Health (CBH), KTH Royal Institute of Technology, Stockholm, Sweden
| | - Petra Hååg
- Department of Oncology-Pathology, Karolinska Institutet, 17164, Stockholm, Sweden
| | - Rolf Lewensohn
- Department of Oncology-Pathology, Karolinska Institutet, 17164, Stockholm, Sweden; Theme Cancer, Medical Unit Head and Neck, Lung, and Skin Tumors, Thoracic Oncology Center, Karolinska University Hospital, S-171 64, Solna, Sweden
| | - Kristina Viktorsson
- Department of Oncology-Pathology, Karolinska Institutet, 17164, Stockholm, Sweden
| | - Amelie E Karlström
- Department of Protein Science, School of Chemistry, Biotechnology, and Health (CBH), KTH Royal Institute of Technology, Stockholm, Sweden
| | - Apurba Dev
- Department of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, 10691, Stockholm, Sweden; Division of Solid-State Electronics, Department of Electrical Engineering, Uppsala University, 75121, Uppsala, Sweden.
| |
Collapse
|
20
|
Stridfeldt F, Cavallaro S, Hååg P, Lewensohn R, Linnros J, Viktorsson K, Dev A. Analyses of single extracellular vesicles from non-small lung cancer cells to reveal effects of epidermal growth factor receptor inhibitor treatments. Talanta 2023; 259:124553. [PMID: 37084607 DOI: 10.1016/j.talanta.2023.124553] [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: 11/15/2022] [Revised: 03/29/2023] [Accepted: 04/11/2023] [Indexed: 04/23/2023]
Abstract
Precision cancer medicine has changed the treatment landscape of non-small cell lung cancer (NSCLC) as illustrated by the introduction of tyrosine kinase inhibitors (TKIs) towards mutated epidermal growth factor receptor (EGFR). However, as responses to EGFR-TKIs are heterogenous among NSCLC patients, there is a need for ways to early monitor changes in treatment response in a non-invasive way e.g., in patient's blood samples. Recently, extracellular vesicles (EVs) have been identified as a source of tumor biomarkers which could improve on non-invasive liquid biopsy-based diagnosis of cancer. However, the heterogeneity in EVs is high. Putative biomarker candidates may be hidden in the differential expression of membrane proteins in a subset of EVs hard to identify using bulk techniques. Using a fluorescence-based approach, we demonstrate that a single-EV technique can detect alterations in EV surface protein profiles. We analyzed EVs isolated from an EGFR-mutant NSCLC cell line, which is refractory to EGFR-TKIs erlotinib and responsive to osimertinib, before and after treatment with these drugs and after cisplatin chemotherapy. We studied expression level of five proteins; two tetraspanins (CD9, CD81), and three markers of interest in lung cancer (EGFR, programmed death-ligand 1 (PD-L1), human epidermal growth factor receptor 2 (HER2)). The data reveal alterations induced by the osimertinib treatment compared to the other two treatments. These include the growth of the PD-L1/HER2-positive EV population, with the largest increase in vesicles exclusively expressing one of the two proteins. The expression level per EV decreased for these markers. On the other hand, both the TKIs had a similar effect on the EGFR-positive EV population.
Collapse
Affiliation(s)
- Fredrik Stridfeldt
- Department of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, 10691, Stockholm, Sweden.
| | - Sara Cavallaro
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - Petra Hååg
- Department of Oncology-Pathology, Karolinska Institutet, 171 64, Solna, Sweden
| | - Rolf Lewensohn
- Department of Oncology-Pathology, Karolinska Institutet, 171 64, Solna, Sweden; Theme Cancer, Medical Unit head and neck, lung, and skin tumors, Thoracic Oncology Center, Karolinska University Hospital, SE-171 64, Solna, Sweden
| | - Jan Linnros
- Department of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, 10691, Stockholm, Sweden
| | - Kristina Viktorsson
- Department of Oncology-Pathology, Karolinska Institutet, 171 64, Solna, Sweden
| | - Apurba Dev
- Department of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, 10691, Stockholm, Sweden; Department of Electrical Engineering, Ångströmslaboratoriet, Uppsala University, Uppsala, Box 534, SE-751-21, Sweden.
| |
Collapse
|
21
|
Puthukodan S, Hofmann M, Mairhofer M, Janout H, Schurr J, Hauser F, Naderer C, Preiner J, Winkler S, Sivun D, Jacak J. Purification Analysis, Intracellular Tracking, and Colocalization of Extracellular Vesicles Using Atomic Force and 3D Single-Molecule Localization Microscopy. Anal Chem 2023; 95:6061-6070. [PMID: 37002540 PMCID: PMC10100414 DOI: 10.1021/acs.analchem.3c00144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023]
Abstract
Extracellular vesicles (EVs) play a key role in cell-cell communication and thus have great potential to be utilized as therapeutic agents and diagnostic tools. In this study, we implemented single-molecule microscopy techniques as a toolbox for a comprehensive characterization as well as measurement of the cellular uptake of HEK293T cell-derived EVs (eGFP-labeled) in HeLa cells. A combination of fluorescence and atomic force microscopy revealed a fraction of 68% fluorescently labeled EVs with an average size of ∼45 nm. Two-color single-molecule fluorescence microscopy analysis elucidated the 3D dynamics of EVs entering HeLa cells. 3D colocalization analysis of two-color direct stochastic optical reconstruction microscopy (dSTORM) images revealed that 25% of EVs that experienced uptake colocalized with transferrin, which has been linked to early recycling of endosomes and clathrin-mediated endocytosis. The localization analysis was combined with stepwise photobleaching, providing a comparison of protein aggregation outside and inside the cells.
Collapse
Affiliation(s)
| | - Martina Hofmann
- University
of Applied Sciences Upper Austria, Linz 4020, Austria
| | - Mario Mairhofer
- University
of Applied Sciences Upper Austria, Linz 4020, Austria
| | - Hannah Janout
- University
of Applied Sciences Upper Austria, Hagenberg 4232, Austria
- Department
of Computer Science, Johannes Kepler University, Linz 4040, Austria
| | - Jonas Schurr
- University
of Applied Sciences Upper Austria, Hagenberg 4232, Austria
- Department
of Computer Science, Johannes Kepler University, Linz 4040, Austria
| | - Fabian Hauser
- University
of Applied Sciences Upper Austria, Linz 4020, Austria
| | | | - Johannes Preiner
- University
of Applied Sciences Upper Austria, Linz 4020, Austria
| | - Stephan Winkler
- University
of Applied Sciences Upper Austria, Hagenberg 4232, Austria
- Department
of Computer Science, Johannes Kepler University, Linz 4040, Austria
| | - Dmitry Sivun
- University
of Applied Sciences Upper Austria, Linz 4020, Austria
| | - Jaroslaw Jacak
- University
of Applied Sciences Upper Austria, Linz 4020, Austria
- AUVA
Research Center, Ludwig Boltzmann Institute
for Experimental and Clinical Traumatology, Vienna 1200, Austria
| |
Collapse
|
22
|
Feng Y, Liu M, Li X, Li M, Xing X, Liu L. Nanomechanical Signatures of Extracellular Vesicles from Hematologic Cancer Patients Unraveled by Atomic Force Microscopy for Liquid Biopsy. NANO LETTERS 2023; 23:1591-1599. [PMID: 36723485 DOI: 10.1021/acs.nanolett.3c00093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Cells release extracellular vesicles (EVs) as the carriers for intercellular communications to regulate life activities. Particularly, it is increasingly apparent that mechanical forces play an essential role in biological systems. The nanomechanical properties of EVs and their dynamics in cancer development are still not fully understood. Herein, with the use of atomic force microscopy (AFM), the nanomechanical signatures of EVs from the liquid biopsies of hematologic cancer patients were unraveled. Single native EVs were probed by AFM under aqueous conditions. The elastic and viscous properties of EVs were measured and visualized to correlate EV mechanics with EV geometry. Experimental results remarkably reveal the significant differences in EV mechanics among multiple myeloma patients, lymphoma patients, and healthy volunteers. The study unveils the unique nanomechanical signatures of EVs in hematologic cancers, which will benefit the studies of liquid biopsies for cancer diagnosis and prognosis with translational significance.
Collapse
Affiliation(s)
- Yaqi Feng
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, People's Republic of China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Meichen Liu
- Cancer Hospital of China Medical University, Shenyang 110042, People's Republic of China
- Liaoning Cancer Hospital & Institute, Shenyang 110042, People's Republic of China
| | - Xinxin Li
- Cancer Hospital of China Medical University, Shenyang 110042, People's Republic of China
- Liaoning Cancer Hospital & Institute, Shenyang 110042, People's Republic of China
| | - Mi Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, People's Republic of China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Xiaojing Xing
- Cancer Hospital of China Medical University, Shenyang 110042, People's Republic of China
- Liaoning Cancer Hospital & Institute, Shenyang 110042, People's Republic of China
| | - Lianqing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, People's Republic of China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
23
|
Chin AR. Challenges for Studying and Isolating Extracellular Vesicles from Cell-Conditioned Media. Methods Mol Biol 2023; 2666:299-315. [PMID: 37166673 DOI: 10.1007/978-1-0716-3191-1_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Extracellular vesicles (EV) are small (100-1000 nm) particles that cells release into the extracellular space that have become increasingly famous for their potential in regenerative medicine and for their alterations in diseases such as cancer to promote disease progression, in particular for their potential for intercellular communication. However, studying EV can be challenging due to the broad diversity of both the EV themselves as well as the methods used to study them. This chapter aims to help investigators new to the EV field by describing challenges with studying EV, methods for enriching EV, and a simple EV enrichment protocol using differential ultracentrifugation.
Collapse
Affiliation(s)
- Andrew R Chin
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA.
| |
Collapse
|
24
|
De Stefano N, Calleri A, Navarro-Tableros V, Rigo F, Patrono D, Romagnoli R. State-of-the-Art and Future Directions in Organ Regeneration with Mesenchymal Stem Cells and Derived Products during Dynamic Liver Preservation. Medicina (B Aires) 2022; 58:medicina58121826. [PMID: 36557029 PMCID: PMC9785426 DOI: 10.3390/medicina58121826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/29/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Transplantation is currently the treatment of choice for end-stage liver diseases but is burdened by the shortage of donor organs. Livers from so-called extended-criteria donors represent a valid option to overcome organ shortage, but they are at risk for severe post-operative complications, especially when preserved with conventional static cold storage. Machine perfusion technology reduces ischemia-reperfusion injury and allows viability assessment of these organs, limiting their discard rate and improving short- and long-term outcomes after transplantation. Moreover, by keeping the graft metabolically active, the normothermic preservation technique guarantees a unique platform to administer regenerative therapies ex vivo. With their anti-inflammatory and immunomodulatory properties, mesenchymal stem cells are among the most promising sources of therapies for acute and chronic liver failure, but their routine clinical application is limited by several biosafety concerns. It is emerging that dynamic preservation and stem cell therapy may supplement each other if combined, as machine perfusion can be used to deliver stem cells to highly injured grafts, avoiding potential systemic side effects. The aim of this narrative review is to provide a comprehensive overview on liver preservation techniques and mesenchymal stem cell-based therapies, focusing on their application in liver graft reconditioning.
Collapse
Affiliation(s)
- Nicola De Stefano
- General Surgery 2U—Liver Transplant Unit, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Torino, 10126 Turin, Italy
| | - Alberto Calleri
- Gastrohepatology Unit, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Torino, 10126 Turin, Italy
| | - Victor Navarro-Tableros
- 2i3T, Società per la Gestione dell’incubatore di Imprese e per il Trasferimento Tecnologico, University of Torino, 10126 Turin, Italy
| | - Federica Rigo
- General Surgery 2U—Liver Transplant Unit, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Torino, 10126 Turin, Italy
| | - Damiano Patrono
- General Surgery 2U—Liver Transplant Unit, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Torino, 10126 Turin, Italy
| | - Renato Romagnoli
- General Surgery 2U—Liver Transplant Unit, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Torino, 10126 Turin, Italy
- Correspondence: ; Tel.: +39-011-6334364
| |
Collapse
|
25
|
Jin S, Lv Z, Kang L, Wang J, Tan C, Shen L, Wang L, Liu J. Next generation of neurological therapeutics: Native and bioengineered extracellular vesicles derived from stem cells. Asian J Pharm Sci 2022; 17:779-797. [PMID: 36600903 PMCID: PMC9800941 DOI: 10.1016/j.ajps.2022.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/20/2022] [Accepted: 10/10/2022] [Indexed: 11/19/2022] Open
Abstract
Extracellular vesicles (EVs)-based cell-free therapy, particularly stem cell-derived extracellular vesicles (SC-EVs), offers new insights into treating a series of neurological disorders and becomes a promising candidate for alternative stem cell regenerative therapy. Currently, SC-EVs are considered direct therapeutic agents by themselves and/or dynamic delivery systems as they have a similar regenerative capacity of stem cells to promote neurogenesis and can easily load many functional small molecules to recipient cells in the central nervous system. Meanwhile, as non-living entities, SC-EVs avoid the uncontrollability and manufacturability limitations of live stem cell products in vivo (e.g., low survival rate, immune response, and tumorigenicity) and in vitro (e.g., restricted sources, complex preparation processes, poor quality control, low storage, shipping instability, and ethical controversy) by strict quality control system. Moreover, SC-EVs can be engineered or designed to enhance further overall yield, increase bioactivity, improve targeting, and extend their half-life. Here, this review provides an overview on the biological properties of SC-EVs, and the current progress in the strategies of native or bioengineered SC-EVs for nerve injury repairing is presented. Then we further summarize the challenges of recent research and perspectives for successful clinical application to advance SC-EVs from bench to bedside in neurological diseases.
Collapse
Affiliation(s)
- Shilin Jin
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China
| | - Zhongyue Lv
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China
| | - Lin Kang
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China
| | - Jiayi Wang
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China
| | - Chengcheng Tan
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China
| | - Liming Shen
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China
| | - Liang Wang
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China
| | - Jing Liu
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China
| |
Collapse
|
26
|
Banijamali M, Höjer P, Nagy A, Hååg P, Gomero EP, Stiller C, Kaminskyy VO, Ekman S, Lewensohn R, Karlström AE, Viktorsson K, Ahmadian A. Characterizing single extracellular vesicles by droplet barcode sequencing for protein analysis. J Extracell Vesicles 2022; 11:e12277. [DOI: 10.1002/jev2.12277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 09/29/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Mahsan Banijamali
- Royal Institute of Technology (KTH), School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Gene Technology Science for Life Laboratory Solna Sweden
| | - Pontus Höjer
- Royal Institute of Technology (KTH), School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Gene Technology Science for Life Laboratory Solna Sweden
| | - Abel Nagy
- Royal Institute of Technology (KTH), School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Protein Science AlbaNova University Center Stockholm Sweden
| | - Petra Hååg
- Department of Oncology‐Pathology Karolinska Institutet Solna Sweden
| | - Elizabeth Paz Gomero
- Royal Institute of Technology (KTH), School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Protein Science AlbaNova University Center Stockholm Sweden
| | - Christiane Stiller
- Royal Institute of Technology (KTH), School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Protein Science AlbaNova University Center Stockholm Sweden
| | - Vitaliy O. Kaminskyy
- Department of Oncology‐Pathology Karolinska Institutet Solna Sweden
- Department of Physiology and Pharmacology Karolinska Institutet Stockholm Sweden
| | - Simon Ekman
- Department of Oncology‐Pathology Karolinska Institutet Solna Sweden
- Theme Cancer, Medical Unit head and Neck, Lung, and Skin Tumors, Thoracic Oncology Center Karolinska University Hospital Solna Sweden
| | - Rolf Lewensohn
- Department of Oncology‐Pathology Karolinska Institutet Solna Sweden
- Theme Cancer, Medical Unit head and Neck, Lung, and Skin Tumors, Thoracic Oncology Center Karolinska University Hospital Solna Sweden
| | - Amelie Eriksson Karlström
- Royal Institute of Technology (KTH), School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Protein Science AlbaNova University Center Stockholm Sweden
| | | | - Afshin Ahmadian
- Royal Institute of Technology (KTH), School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Gene Technology Science for Life Laboratory Solna Sweden
| |
Collapse
|
27
|
Bai J, Wei X, Zhang X, Wu C, Wang Z, Chen M, Wang J. Microfluidic strategies for the isolation and profiling of exosomes. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
28
|
Görgens A, Corso G, Hagey DW, Jawad Wiklander R, Gustafsson MO, Felldin U, Lee Y, Bostancioglu RB, Sork H, Liang X, Zheng W, Mohammad DK, van de Wakker SI, Vader P, Zickler AM, Mamand DR, Ma L, Holme MN, Stevens MM, Wiklander OPB, EL Andaloussi S. Identification of storage conditions stabilizing extracellular vesicles preparations. J Extracell Vesicles 2022; 11:e12238. [PMID: 35716060 PMCID: PMC9206228 DOI: 10.1002/jev2.12238] [Citation(s) in RCA: 177] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/23/2022] [Accepted: 05/16/2022] [Indexed: 12/31/2022] Open
Abstract
Extracellular vesicles (EVs) play a key role in many physiological and pathophysiological processes and hold great potential for therapeutic and diagnostic use. Despite significant advances within the last decade, the key issue of EV storage stability remains unresolved and under investigated. Here, we aimed to identify storage conditions stabilizing EVs and comprehensively compared the impact of various storage buffer formulations at different temperatures on EVs derived from different cellular sources for up to 2 years. EV features including concentration, diameter, surface protein profile and nucleic acid contents were assessed by complementary methods, and engineered EVs containing fluorophores or functionalized surface proteins were utilized to compare cellular uptake and ligand binding. We show that storing EVs in PBS over time leads to drastically reduced recovery particularly for pure EV samples at all temperatures tested, starting already within days. We further report that using PBS as diluent was found to result in severely reduced EV recovery rates already within minutes. Several of the tested new buffer conditions largely prevented the observed effects, the lead candidate being PBS supplemented with human albumin and trehalose (PBS-HAT). We report that PBS-HAT buffer facilitates clearly improved short-term and long-term EV preservation for samples stored at -80°C, stability throughout several freeze-thaw cycles, and drastically improved EV recovery when using a diluent for EV samples for downstream applications.
Collapse
Affiliation(s)
- André Görgens
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
- Institute for Transfusion MedicineUniversity Hospital Essen, University of Duisburg‐EssenEssenGermany
- Evox Therapeutics LimitedOxfordUK
| | - Giulia Corso
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Daniel W. Hagey
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Rim Jawad Wiklander
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Manuela O. Gustafsson
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Ulrika Felldin
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Yi Lee
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - R. Beklem Bostancioglu
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Helena Sork
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
- Institute of TechnologyUniversity of TartuTartuEstonia
| | - Xiuming Liang
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Wenyi Zheng
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Dara K. Mohammad
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
- College of Agricultural Engineering SciencesSalahaddin University‐ErbilErbilKurdistan RegionIraq
| | - Simonides I. van de Wakker
- Department of CardiologyExperimental Cardiology LaboratoryUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Pieter Vader
- Department of CardiologyExperimental Cardiology LaboratoryUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
- CDL ResearchUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Antje M. Zickler
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Doste R. Mamand
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Li Ma
- Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Margaret N. Holme
- Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Molly M. Stevens
- Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
- Department of MaterialsDepartment of Bioengineeringand Institute of Biomedical EngineeringImperial College LondonLondonUK
| | - Oscar P. B. Wiklander
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
- Evox Therapeutics LimitedOxfordUK
| | - Samir EL Andaloussi
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
- Evox Therapeutics LimitedOxfordUK
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| |
Collapse
|
29
|
Extracellular Vesicles as Novel Drug-Delivery Systems through Intracellular Communications. MEMBRANES 2022; 12:membranes12060550. [PMID: 35736256 PMCID: PMC9230693 DOI: 10.3390/membranes12060550] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 02/07/2023]
Abstract
Since it has been reported that extracellular vesicles (EVs) carry cargo using cell-to-cell comminication according to various in vivo situations, they are exprected to be applied as new drug-delivery systems (DDSs). In addition, non-coding RNAs, such as microRNAs (miRNAs), have attracted much attention as potential biomarkers in the encapsulated extracellular-vesicle (EV) form. EVs are bilayer-based lipids with heterogeneous populations of varying sizes and compositions. The EV-mediated transport of contents, which includes proteins, lipids, and nucleic acids, has attracted attention as a DDS through intracellular communication. Many reports have been made on the development of methods for introducing molecules into EVs and efficient methods for introducing them into target vesicles. In this review, we outline the possible molecular mechanisms by which miRNAs in exosomes participate in the post-transcriptional regulation of signaling pathways via cell–cell communication as novel DDSs, especially small EVs.
Collapse
|
30
|
Wang T, Xing Y, Cheng Z, Yu F. Analysis of Single Extracellular Vesicles for Biomedical Applications with Especial Emphasis on Cancer Investigations. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
31
|
Wang L, Zhu Z, Liao Y, Zhang L, Yu Z, Yang R, Wu J, Wu Z, Sun X. Host Liver-Derived Extracellular Vesicles Deliver miR-142a-3p Induces Neutrophil Extracellular Traps via Targeting WASL to Block the Development of Schistosoma japonicum. Mol Ther 2022; 30:2092-2107. [PMID: 35351657 PMCID: PMC9092393 DOI: 10.1016/j.ymthe.2022.03.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/10/2022] [Accepted: 03/24/2022] [Indexed: 10/18/2022] Open
Abstract
Schistosomiasis is an important neglected tropical disease. Interactions between the host immune system and schistosomes are complex. Neutrophils contribute to clearance of large pathogens primarily by releasing neutrophil extracellular traps (NETs). However, the functional role of NETs in clearing schistosomes remains unclear. Herein, we report that extracellular vesicles (EVs) derived from the liver of Schistosoma japonicum-infected mice (IL-EVs) induce NET release by delivering miR-142a-3p to target WASL and block the development of S. japonicum. WASL knockout accelerated the formation of NETs that blocked further development of S. japonicum. miR-142a-3p and NETs upregulated the expression of CCL2, which recruits macrophages that block S. japonicum development. However, S. japonicum inhibited NET formation in wild-type mice by upregulating host interleukin-10 (IL-10) expression. In contrast, in WASL knockout mice, IL-10 expression was downregulated, and S. japonicum-mediated inhibition of NET formation was significantly reduced. IL-EV-mediated induction of NET formation is thus an anti-schistosome response that can be counteracted by S. japonicum. These findings suggest that IL-EV-mediated induction of NET formation plays a key role in schistosome infection and that WASL is a potential therapeutic target in schistosomiasis and other infectious diseases.
Collapse
|
32
|
Jiang C, Fu Y, Liu G, Shu B, Davis J, Tofaris GK. Multiplexed Profiling of Extracellular Vesicles for Biomarker Development. NANO-MICRO LETTERS 2021; 14:3. [PMID: 34855021 PMCID: PMC8638654 DOI: 10.1007/s40820-021-00753-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/22/2021] [Indexed: 05/09/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived membranous particles that play a crucial role in molecular trafficking, intercellular transport and the egress of unwanted proteins. They have been implicated in many diseases including cancer and neurodegeneration. EVs are detected in all bodily fluids, and their protein and nucleic acid content offers a means of assessing the status of the cells from which they originated. As such, they provide opportunities in biomarker discovery for diagnosis, prognosis or the stratification of diseases as well as an objective monitoring of therapies. The simultaneous assaying of multiple EV-derived markers will be required for an impactful practical application, and multiplexing platforms have evolved with the potential to achieve this. Herein, we provide a comprehensive overview of the currently available multiplexing platforms for EV analysis, with a primary focus on miniaturized and integrated devices that offer potential step changes in analytical power, throughput and consistency.
Collapse
Affiliation(s)
- Cheng Jiang
- Nuffield Department of Clinical Neurosciences, New Biochemistry Building, University of Oxford, Oxford, OX1 3QU, UK.
- Department of Chemistry, University of Oxford, Oxford, OX1 3QZ, UK.
- Kavli Institute for Nanoscience Discovery, New Biochemistry Building, University of Oxford, Oxford, UK.
| | - Ying Fu
- Department of Chemistry, University of Oxford, Oxford, OX1 3QZ, UK
| | - Guozhen Liu
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, 518172, People's Republic of China
| | - Bowen Shu
- Dermatology Hospital, Southern Medical University, Guangzhou, 510091, People's Republic of China
| | - Jason Davis
- Department of Chemistry, University of Oxford, Oxford, OX1 3QZ, UK.
| | - George K Tofaris
- Nuffield Department of Clinical Neurosciences, New Biochemistry Building, University of Oxford, Oxford, OX1 3QU, UK.
- Kavli Institute for Nanoscience Discovery, New Biochemistry Building, University of Oxford, Oxford, UK.
| |
Collapse
|
33
|
Exosomes in nasopharyngeal carcinoma. Clin Chim Acta 2021; 523:355-364. [PMID: 34666030 DOI: 10.1016/j.cca.2021.10.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 12/18/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant epithelial tumor with a unique geographical distribution, primarily prevalent in East Africa and Asia. Although there is an increased understanding of the pathogenesis and risk factors of NPC, prevention and treatment efforts remain limited. Various studies have indicated that exosomes are actively involved in NPC by delivering biomolecules such as non-coding RNAs and proteins to target cells. In this review, we summarize the biological functions of exosomes in NPC and highlight their prospects as diagnostic biomarkers. In NPC, exosomes can manipulate the tumor microenvironment, participate in chemotherapy and radiation resistance, induce immune suppression, promote pathological angiogenesis, and support metastasis, and thus they could also be promising biomarkers. Because exosomes have essential effects and unusual biological properties, they have a promising future in diagnostic monitoring and prognostic evaluation. Although there are technical issues associated with using exosomes in large-scale applications, they have unparalleled advantages in assisting the clinical management of NPC.
Collapse
|
34
|
Sahu SS, Cavallaro S, Hååg P, Nagy Á, Karlström AE, Lewensohn R, Viktorsson K, Linnros J, Dev A. Exploiting Electrostatic Interaction for Highly Sensitive Detection of Tumor-Derived Extracellular Vesicles by an Electrokinetic Sensor. ACS APPLIED MATERIALS & INTERFACES 2021; 13:42513-42521. [PMID: 34473477 PMCID: PMC8447189 DOI: 10.1021/acsami.1c13192] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
We present an approach to improve the detection sensitivity of a streaming current-based biosensor for membrane protein profiling of small extracellular vesicles (sEVs). The experimental approach, supported by theoretical investigation, exploits electrostatic charge contrast between the sensor surface and target analytes to enhance the detection sensitivity. We first demonstrate the feasibility of the approach using different chemical functionalization schemes to modulate the zeta potential of the sensor surface in a range -16.0 to -32.8 mV. Thereafter, we examine the sensitivity of the sensor surface across this range of zeta potential to determine the optimal functionalization scheme. The limit of detection (LOD) varied by 2 orders of magnitude across this range, reaching a value of 4.9 × 106 particles/mL for the best performing surface for CD9. We then used the optimized surface to profile CD9, EGFR, and PD-L1 surface proteins of sEVs derived from non-small cell lung cancer (NSCLC) cell-line H1975, before and after treatment with EGFR tyrosine kinase inhibitors, as well as sEVs derived from pleural effusion fluid of NSCLC adenocarcinoma patients. Our results show the feasibility to monitor CD9, EGFR, and PD-L1 expression on the sEV surface, illustrating a good prospect of the method for clinical application.
Collapse
Affiliation(s)
- Siddharth Sourabh Sahu
- Department
of Electrical Engineering, The Ångström Laboratory, Uppsala University, 75121 Uppsala, Sweden
| | - Sara Cavallaro
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, 10691 Stockholm, Sweden
| | - Petra Hååg
- Department
of Oncology-Pathology, Karolinska Institutet, 17164 Stockholm, Sweden
| | - Ábel Nagy
- Department
of Protein Science, School of Chemistry, Biotechnology, and Health
(CBH), KTH Royal Institute of Technology, 10691 Stockholm, Sweden
| | - Amelie Eriksson Karlström
- Department
of Protein Science, School of Chemistry, Biotechnology, and Health
(CBH), KTH Royal Institute of Technology, 10691 Stockholm, Sweden
| | - Rolf Lewensohn
- Department
of Oncology-Pathology, Karolinska Institutet, 17164 Stockholm, Sweden
- Theme
Cancer, Patient Area Head and Neck, Lung, and Skin, Karolinska University Hospital, 17164 Solna, Sweden
| | - Kristina Viktorsson
- Department
of Oncology-Pathology, Karolinska Institutet, 17164 Stockholm, Sweden
| | - Jan Linnros
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, 10691 Stockholm, Sweden
| | - Apurba Dev
- Department
of Electrical Engineering, The Ångström Laboratory, Uppsala University, 75121 Uppsala, Sweden
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, 10691 Stockholm, Sweden
| |
Collapse
|
35
|
Cavallaro S, Hååg P, Viktorsson K, Krozer A, Fogel K, Lewensohn R, Linnros J, Dev A. Comparison and optimization of nanoscale extracellular vesicle imaging by scanning electron microscopy for accurate size-based profiling and morphological analysis. NANOSCALE ADVANCES 2021; 3:3053-3063. [PMID: 36133670 PMCID: PMC9419097 DOI: 10.1039/d0na00948b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/21/2021] [Indexed: 05/05/2023]
Abstract
Nanosized extracellular vesicles (EVs) have been found to play a key role in intercellular communication, offering opportunities for both disease diagnostics and therapeutics. However, lying below the diffraction limit and also being highly heterogeneous in their size, morphology and abundance, these vesicles pose significant challenges for physical characterization. Here, we present a direct visual approach for their accurate morphological and size-based profiling by using scanning electron microscopy (SEM). To achieve that, we methodically examined various process steps and developed a protocol to improve the throughput, conformity and image quality while preserving the shape of EVs. The study was performed with small EVs (sEVs) isolated from a non-small-cell lung cancer (NSCLC) cell line as well as from human serum, and the results were compared with those obtained from nanoparticle tracking analysis (NTA). While the comparison of the sEV size distributions showed good agreement between the two methods for large sEVs (diameter > 70 nm), the microscopy based approach showed a better capacity for analyses of smaller vesicles, with higher sEV counts compared to NTA. In addition, we demonstrated the possibility of identifying non-EV particles based on size and morphological features. The study also showed process steps that can generate artifacts bearing resemblance with sEVs. The results therefore present a simple way to use a widely available microscopy tool for accurate and high throughput physical characterization of EVs.
Collapse
Affiliation(s)
- Sara Cavallaro
- Department of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology 10691 Stockholm Sweden
| | - Petra Hååg
- Department of Oncology-Pathology, Karolinska Institutet 17164 Solna Sweden
| | | | - Anatol Krozer
- Department of Smart Hardware, Division of Digital Systems, Research Institutes of Sweden AB 40014 Gothenburg Sweden
| | - Kristina Fogel
- Department of Smart Hardware, Division of Digital Systems, Research Institutes of Sweden AB 40014 Gothenburg Sweden
| | - Rolf Lewensohn
- Department of Oncology-Pathology, Karolinska Institutet 17164 Solna Sweden
- Theme Cancer, Medical Unit Head and Neck, Lung, and Skin Tumors, Thoracic Oncology Center, Karolinska University Hospital 17164 Solna Sweden
| | - Jan Linnros
- Department of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology 10691 Stockholm Sweden
| | - Apurba Dev
- Department of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology 10691 Stockholm Sweden
- Department of Electrical Engineering, The Ångström Laboratory, Uppsala University 75121 Uppsala Sweden
| |
Collapse
|
36
|
Malenica M, Vukomanović M, Kurtjak M, Masciotti V, dal Zilio S, Greco S, Lazzarino M, Krušić V, Perčić M, Jelovica Badovinac I, Wechtersbach K, Vidović I, Baričević V, Valić S, Lučin P, Kojc N, Grabušić K. Perspectives of Microscopy Methods for Morphology Characterisation of Extracellular Vesicles from Human Biofluids. Biomedicines 2021; 9:603. [PMID: 34073297 PMCID: PMC8228884 DOI: 10.3390/biomedicines9060603] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/18/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are nanometric membranous structures secreted from almost every cell and present in biofluids. Because EV composition reflects the state of its parental tissue, EVs possess an enormous diagnostic/prognostic potential to reveal pathophysiological conditions. However, a prerequisite for such usage of EVs is their detailed characterisation, including visualisation which is mainly achieved by atomic force microscopy (AFM) and electron microscopy (EM). Here we summarise the EV preparation protocols for AFM and EM bringing out the main challenges in the imaging of EVs, both in their natural environment as biofluid constituents and in a saline solution after EV isolation. In addition, we discuss approaches for EV imaging and identify the potential benefits and disadvantages when different AFM and EM methods are applied, including numerous factors that influence the morphological characterisation, standardisation, or formation of artefacts. We also demonstrate the effects of some of these factors by using cerebrospinal fluid as an example of human biofluid with a simpler composition. Here presented comparison of approaches to EV imaging should help to estimate the current state in morphology research of EVs from human biofluids and to identify the most efficient pathways towards the standardisation of sample preparation and microscopy modes.
Collapse
Affiliation(s)
- Mladenka Malenica
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, HR-51000 Rijeka, Croatia; (V.K.); (P.L.); (K.G.)
| | - Marija Vukomanović
- Advanced Materials Department, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia; (M.V.); (M.K.)
| | - Mario Kurtjak
- Advanced Materials Department, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia; (M.V.); (M.K.)
| | - Valentina Masciotti
- CNR-IOM Istituto Officina dei Materiali-Consiglio Nazionale delle Ricerche c/Area Scinece Park, Basovizza, I-34149 Trieste, Italy; (V.M.); (S.d.Z.); (M.L.)
| | - Simone dal Zilio
- CNR-IOM Istituto Officina dei Materiali-Consiglio Nazionale delle Ricerche c/Area Scinece Park, Basovizza, I-34149 Trieste, Italy; (V.M.); (S.d.Z.); (M.L.)
| | | | - Marco Lazzarino
- CNR-IOM Istituto Officina dei Materiali-Consiglio Nazionale delle Ricerche c/Area Scinece Park, Basovizza, I-34149 Trieste, Italy; (V.M.); (S.d.Z.); (M.L.)
| | - Vedrana Krušić
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, HR-51000 Rijeka, Croatia; (V.K.); (P.L.); (K.G.)
| | - Marko Perčić
- Faculty of Engineering, University of Rijeka, HR-51000 Rijeka, Croatia;
- Centre for Micro- and Nanosciences and Technologies, University of Rijeka, HR-51000 Rijeka, Croatia;
| | - Ivana Jelovica Badovinac
- Centre for Micro- and Nanosciences and Technologies, University of Rijeka, HR-51000 Rijeka, Croatia;
- Department of Physics, University of Rijeka, HR-51000 Rijeka, Croatia
| | - Karmen Wechtersbach
- Faculty of Medicine, Institute of Pathology, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (K.W.); (N.K.)
| | - Ivona Vidović
- Department of Biotechnology, University of Rijeka, HR-51000 Rijeka, Croatia; (I.V.); (V.B.)
| | - Vanja Baričević
- Department of Biotechnology, University of Rijeka, HR-51000 Rijeka, Croatia; (I.V.); (V.B.)
| | - Srećko Valić
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, HR-51000 Rijeka, Croatia;
- Division of Physical Chemistry, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| | - Pero Lučin
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, HR-51000 Rijeka, Croatia; (V.K.); (P.L.); (K.G.)
| | - Nika Kojc
- Faculty of Medicine, Institute of Pathology, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (K.W.); (N.K.)
| | - Kristina Grabušić
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, HR-51000 Rijeka, Croatia; (V.K.); (P.L.); (K.G.)
| |
Collapse
|