1
|
Knab F, Guaitoli G, Jarboui MA, von Zweydorf F, Isik FB, Klose F, Rajkumar AP, Gasser T, Gloeckner CJ. The cellular and extracellular proteomic signature of human dopaminergic neurons carrying the LRRK2 G2019S mutation. Front Neurosci 2024; 18:1502246. [PMID: 39726830 PMCID: PMC11669673 DOI: 10.3389/fnins.2024.1502246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
Background Extracellular vesicles are easily accessible in various biofluids and allow the assessment of disease-related changes in the proteome. This has made them a promising target for biomarker studies, especially in the field of neurodegeneration where access to diseased tissue is very limited. Genetic variants in the LRRK2 gene have been linked to both familial and sporadic forms of Parkinson's disease. With LRRK2 inhibitors entering clinical trials, there is an unmet need for biomarkers that reflect LRRK2-specific pathology and target engagement. Methods In this study, we used induced pluripotent stem cells derived from a patient with Parkinson's disease carrying the LRRK2 G2019S mutation and an isogenic gene-corrected control to generate human dopaminergic neurons. We isolated extracellular vesicles and neuronal cell lysates and characterized their proteomic signature using data-independent acquisition proteomics. Then, we performed differential expression analysis to identify dysregulated proteins in the mutated line. We used Metascape and gene ontology enrichment analysis on the dysregulated proteomes to identify changes in associated functional networks. Results We identified 595 significantly differentially regulated proteins in extracellular vesicles and 3,205 in cell lysates. We visualized functionally relevant protein-protein interaction networks and identified key regulators within the dysregulated proteomes. Using gene ontology, we found a close association with biological processes relevant to neurodegeneration and Parkinson's disease. Finally, we focused on proteins that were dysregulated in both the extracellular and cellular proteomes. We provide a list of ten biomarker candidates that are functionally relevant to neurodegeneration and linked to LRRK2-associated pathology, for example, the sonic hedgehog signaling molecule, a protein that has tightly been linked to LRRK2-related disruption of cilia function. Conclusion In conclusion, we characterized the cellular and extracellular proteome of dopaminergic neurons carrying the LRRK2 G2019S mutation and proposed an experimentally based list of biomarker candidates for future studies.
Collapse
Affiliation(s)
- Felix Knab
- Hertie Institute for Clinical Brain Research, Department of Neurodegeneration, University of Tübingen, Tübingen, Germany
| | | | - Mohamed Ali Jarboui
- Core Facility for Medical Proteomics, Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | | | - Fatma Busra Isik
- Institute of Mental Health, Mental Health and Clinical Neurosciences Academic Unit, University of Nottingham, Nottingham, United Kingdom
| | - Franziska Klose
- Core Facility for Medical Proteomics, Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Anto Praveen Rajkumar
- Core Facility for Medical Proteomics, Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Thomas Gasser
- Hertie Institute for Clinical Brain Research, Department of Neurodegeneration, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | | |
Collapse
|
2
|
Nix C, Sulejman S, Fillet M. Development of complementary analytical methods to characterize extracellular vesicles. Anal Chim Acta 2024; 1329:343171. [PMID: 39396273 DOI: 10.1016/j.aca.2024.343171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Extracellular vesicles (EVs) are involved in intercellular communication and various biological processes. They hold clinical promise for the diagnosis and management of a wide range of pathologies, including cancer, cardiovascular diseases and degenerative diseases, and are of interest as regenerative therapies. Understanding the complex structure of these EVs is essential to perceive the current challenges associated with their analysis and characterization. Today, challenges remain in terms of access to high-yield, high-purity isolation methods, as well as analytical methods for characterizing and controlling the quality of these products for clinical use. RESULTS We isolated EVs from the same immortalized human cell culture supernatant using two commonly used approaches, namely differential ultracentrifugation and membrane affinity. Then we evaluated EV morphology, size, zeta potential, particle and protein content, as well as protein identity using cryogenic electron microscopy, nanoparticle tracking analysis, asymmetric field flow fractionation (AF4) and size exclusion chromatography (SEC) coupled to multi angle light scattering, bicinchoninic acid assay, electrophoretic light scattering, western blotting and high-resolution mass spectrometry. Compared to membrane affinity isolation, dUC is a more efficient isolation process for obtaining particles with the characteristics expected for EVs and more specifically for exosomes. To validate an isolation process, cryogenic electron microscopy is essential to confirm vesicles with membranes. High resolution mass spectrometry is powerful for understanding the mechanism of action of vesicles. Separative methods, such as AF4 and SEC, are interesting for separating vesicle subpopulations and contaminants. SIGNIFICANCE This study provides a critical assessment of eight different techniques for analyzing EVs, some of which are mandatory for in-depth characterization and deciphering, while others are more appropriate for routine analysis, once the production and isolation process has been validated. The strengths and limitations of the different approaches used are highlighted.
Collapse
Affiliation(s)
- Cindy Nix
- Laboratory for the Analysis of Medicines (LAM), Department of Pharmacy, CIRM, University of Liege, Avenue Hippocrate 15, B36 Tour 4 +3, 4000, Liège, Belgium
| | - Sanije Sulejman
- Laboratory for the Analysis of Medicines (LAM), Department of Pharmacy, CIRM, University of Liege, Avenue Hippocrate 15, B36 Tour 4 +3, 4000, Liège, Belgium
| | - Marianne Fillet
- Laboratory for the Analysis of Medicines (LAM), Department of Pharmacy, CIRM, University of Liege, Avenue Hippocrate 15, B36 Tour 4 +3, 4000, Liège, Belgium.
| |
Collapse
|
3
|
Chong SW, Shen Y, Palomba S, Vigolo D. Nanofluidic Lab-On-A-Chip Systems for Biosensing in Healthcare. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2407478. [PMID: 39491535 DOI: 10.1002/smll.202407478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/21/2024] [Indexed: 11/05/2024]
Abstract
Biosensing plays a vital role in healthcare monitoring, disease detection, and treatment planning. In recent years, nanofluidic technology has been increasingly explored to be developed into lab-on-a-chip biosensing systems. Given now the possibility of fabricating geometrically defined nanometric channels that are commensurate with the size of many biomolecules, nanofluidic-based devices are likely to become a key technology for the analysis of various clinical biomarkers, including DNA (deoxyribonucleic acid) and proteins in liquid biopsies. This review summarizes the fundamentals and technological advances of nanofluidics from the purview of single-molecule analysis, detection of low-abundance molecules, and single-cell analysis at the subcellular level. The extreme confinement and dominant surface charge effects in nanochannels provide unique advantages to nanofluidic devices for the manipulation and transport of target biomarkers. When coupled to a microfluidic network to facilitate sample introduction, integrated micro-nanofluidic biosensing devices are proving to be more sensitive and specific in molecular analysis compared to conventional assays in many cases. Based on recent progress in nanofluidics and current clinical trends, the review concludes with a discussion of near-term challenges and future directions for the development of nanofluidic-based biosensing systems toward enabling a new wave of lab-on-a-chip technology for personalized and preventive medicine.
Collapse
Affiliation(s)
- Shin Wei Chong
- School of Biomedical Engineering, The University of Sydney, Sydney, NSW, 2006, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Yi Shen
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Stefano Palomba
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
- School of Physics, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Daniele Vigolo
- School of Biomedical Engineering, The University of Sydney, Sydney, NSW, 2006, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
4
|
Villa C, Secchi V, Macchi M, Tripodi L, Trombetta E, Zambroni D, Padelli F, Mauri M, Molinaro M, Oddone R, Farini A, De Palma A, Varela Pinzon L, Santarelli F, Simonutti R, Mauri P, Porretti L, Campione M, Aquino D, Monguzzi A, Torrente Y. Magnetic-field-driven targeting of exosomes modulates immune and metabolic changes in dystrophic muscle. NATURE NANOTECHNOLOGY 2024; 19:1532-1543. [PMID: 39039121 PMCID: PMC11486659 DOI: 10.1038/s41565-024-01725-y] [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: 07/14/2023] [Accepted: 06/18/2024] [Indexed: 07/24/2024]
Abstract
Exosomes are promising therapeutics for tissue repair and regeneration to induce and guide appropriate immune responses in dystrophic pathologies. However, manipulating exosomes to control their biodistribution and targeting them in vivo to achieve adequate therapeutic benefits still poses a major challenge. Here we overcome this limitation by developing an externally controlled delivery system for primed annexin A1 myo-exosomes (Exomyo). Effective nanocarriers are realized by immobilizing the Exomyo onto ferromagnetic nanotubes to achieve controlled delivery and localization of Exomyo to skeletal muscles by systemic injection using an external magnetic field. Quantitative muscle-level analyses revealed that macrophages dominate the uptake of Exomyo from these ferromagnetic nanotubes in vivo to synergistically promote beneficial muscle responses in a murine animal model of Duchenne muscular dystrophy. Our findings provide insights into the development of exosome-based therapies for muscle diseases and, in general, highlight the formulation of effective functional nanocarriers aimed at optimizing exosome biodistribution.
Collapse
Grants
- Regione Lombardia (Region of Lombardy)
- Fondazione Telethon (Telethon Foundation)
- RF-2016-02362263 "Multimodal nanotracking for exosome-based therapy in DMD" (theory enhancing) “At the origin of congenital muscular dystrophy: shedding light on the Tdark proteins DPM2 and DPM3”, Bando “Cariplo Telethon Alliance GJC2021” 2022
- Multiomics pRofiling of patient spEcific Models to predict druggable targets in severe neuromuscular rare diseases (REMODEL)”, Unmet Medical Needs, Fondazione Regionale per la Ricerca Biomedica (FRRB), 2022 Nanoparticles in Freidreich Ataxia” National Center for Gene Therapy and Drugs based on RNA Technology, Spoke #1: Genetic diseases, PNRR CN3 RNA, 2022
- “Isolamento di nanoparticelle naturali da utilizzare come agenti anti-infiammatori/anti-fibrotici”, 5X1000, Fondazione Patrimonio e dalla Direzione Scientifica Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico (2022)
- PNRR CN3 RNA, 2022, PNRR project ANTHEM: AdvaNced Technologies for Human-centrEd Medicine - PNC0000003 Spoke #2 – NextGenerationEU RF-2016-02362263 "Multimodal nanotracking for exosome-based therapy in DMD" (theory enhancing)
Collapse
Affiliation(s)
- Chiara Villa
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Valeria Secchi
- Department of Materials Science, University of Milano Bicocca, Milan, Italy
- NANOMIB, Nanomedicine Center, University of Milano Bicocca, Milan, Italy
| | - Mirco Macchi
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Luxembourg Centre for Systems Biomedicine, Department of Biomedical Data Science, Luxembourg City, Luxembourg
| | - Luana Tripodi
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Elena Trombetta
- Flow Cytometry Service, Clinical Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Desiree Zambroni
- Advanced Light and Electron Microscopy Bioimaging Center ALEMBIC, San Raffaele Scientific Institute - OSR, Milan, Italy
| | - Francesco Padelli
- Department of Neuroradiology, IRCCS Foundation Neurological Institute 'Carlo Besta', Milan, Italy
| | - Michele Mauri
- Department of Materials Science, University of Milano Bicocca, Milan, Italy
- NANOMIB, Nanomedicine Center, University of Milano Bicocca, Milan, Italy
| | - Monica Molinaro
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Rebecca Oddone
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Andrea Farini
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Antonella De Palma
- National Research Council of Italy, Proteomics and Metabolomics Unit, Institute for Biomedical Technologies, ITB-CNR, Segrate, Milan, Italy
- Clinical Proteomics Laboratory, ITB-CNR, CNR.Biomics Infrastructure, Elixir, Milan, Italy
| | - Laura Varela Pinzon
- Veterinary Medicine, Department Clinical Sciences, Equine Sciences, Equine Musculoskeletal Biology. Utrecht University, Utrecht, Netherlands
| | - Federica Santarelli
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Roberto Simonutti
- Department of Materials Science, University of Milano Bicocca, Milan, Italy
- NANOMIB, Nanomedicine Center, University of Milano Bicocca, Milan, Italy
| | - PierLuigi Mauri
- National Research Council of Italy, Proteomics and Metabolomics Unit, Institute for Biomedical Technologies, ITB-CNR, Segrate, Milan, Italy
- Clinical Proteomics Laboratory, ITB-CNR, CNR.Biomics Infrastructure, Elixir, Milan, Italy
| | - Laura Porretti
- Flow Cytometry Service, Clinical Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marcello Campione
- NANOMIB, Nanomedicine Center, University of Milano Bicocca, Milan, Italy
- Department of Earth and Environmental Sciences, University of Milano Bicocca, Milano, Italy
| | - Domenico Aquino
- Department of Neuroradiology, IRCCS Foundation Neurological Institute 'Carlo Besta', Milan, Italy
| | - Angelo Monguzzi
- Department of Materials Science, University of Milano Bicocca, Milan, Italy
- NANOMIB, Nanomedicine Center, University of Milano Bicocca, Milan, Italy
| | - Yvan Torrente
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
5
|
Morganti C, Bonora M, Ito K. Metabolism and HSC fate: what NADPH is made for. Trends Cell Biol 2024:S0962-8924(24)00141-7. [PMID: 39054107 DOI: 10.1016/j.tcb.2024.07.003] [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: 05/01/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
Mitochondrial metabolism plays a central role in the regulation of hematopoietic stem cell (HSC) biology. Mitochondrial fatty acid oxidation (FAO) is pivotal in controlling HSC self-renewal and differentiation. Herein, we discuss recent evidence suggesting that NADPH generated in the mitochondria can influence the fate of HSCs. Although NADPH has multiple functions, HSCs show high levels of NADPH that are preferentially used for cholesterol biosynthesis. Endogenous cholesterol supports the biogenesis of extracellular vesicles (EVs), which are essential for maintaining HSC properties. We also highlight the significance of EVs in hematopoiesis through autocrine signaling. Elucidating the mitochondrial NADPH-cholesterol axis as part of the metabolic requirements of healthy HSCs will facilitate the development of new therapies for hematological disorders.
Collapse
Affiliation(s)
- Claudia Morganti
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Oncology and Medicine, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY 10461, USA.
| | - Massimo Bonora
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Oncology and Medicine, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY 10461, USA.
| |
Collapse
|
6
|
Mitrut RE, Stranford DM, DiBiase BN, Chan JM, Bailey MD, Luo M, Harper CS, Meade TJ, Wang M, Leonard JN. HaloTag display enables quantitative single-particle characterisation and functionalisation of engineered extracellular vesicles. J Extracell Vesicles 2024; 13:e12469. [PMID: 38965984 PMCID: PMC11224594 DOI: 10.1002/jev2.12469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 06/01/2024] [Indexed: 07/06/2024] Open
Abstract
Extracellular vesicles (EVs) play key roles in diverse biological processes, transport biomolecules between cells and have been engineered for therapeutic applications. A useful EV bioengineering strategy is to express engineered proteins on the EV surface to confer targeting, bioactivity and other properties. Measuring how incorporation varies across a population of EVs is important for characterising such materials and understanding their function, yet it remains challenging to quantitatively characterise the absolute number of engineered proteins incorporated at single-EV resolution. To address these needs, we developed a HaloTag-based characterisation platform in which dyes or other synthetic species can be covalently and stoichiometrically attached to engineered proteins on the EV surface. To evaluate this system, we employed several orthogonal quantification methods, including flow cytometry and fluorescence microscopy, and found that HaloTag-mediated quantification is generally robust across EV analysis methods. We compared HaloTag-labelling to antibody-labelling of EVs using single vesicle flow cytometry, enabling us to measure the substantial degree to which antibody labelling can underestimate proteins present on an EV. Finally, we demonstrate the use of HaloTag to compare between protein designs for EV bioengineering. Overall, the HaloTag system is a useful EV characterisation tool which complements and expands existing methods.
Collapse
Affiliation(s)
- Roxana E. Mitrut
- Department of Chemical and Biological EngineeringNorthwestern UniversityEvanstonIllinoisUSA
- Center for Synthetic BiologyNorthwestern UniversityEvanstonIllinoisUSA
| | - Devin M. Stranford
- Department of Chemical and Biological EngineeringNorthwestern UniversityEvanstonIllinoisUSA
- Center for Synthetic BiologyNorthwestern UniversityEvanstonIllinoisUSA
| | - Beth N. DiBiase
- Department of Chemical and Biological EngineeringNorthwestern UniversityEvanstonIllinoisUSA
- Center for Synthetic BiologyNorthwestern UniversityEvanstonIllinoisUSA
| | - Jonathan M. Chan
- Department of Chemical and Biological EngineeringNorthwestern UniversityEvanstonIllinoisUSA
| | | | - Minrui Luo
- Department of ChemistryNorthwestern UniversityEvanstonIllinoisUSA
- Chemistry of Life Processes InstituteNorthwestern UniversityEvanstonIllinoisUSA
| | - Clare S. Harper
- Interdisciplinary Biological Sciences Training ProgramNorthwestern UniversityEvanstonIllinoisUSA
| | - Thomas J. Meade
- Department of ChemistryNorthwestern UniversityEvanstonIllinoisUSA
- Chemistry of Life Processes InstituteNorthwestern UniversityEvanstonIllinoisUSA
- Interdisciplinary Biological Sciences Training ProgramNorthwestern UniversityEvanstonIllinoisUSA
- Robert H. Lurie Comprehensive Cancer CenterNorthwestern UniversityEvanstonIllinoisUSA
| | - Muzhou Wang
- Department of Chemical and Biological EngineeringNorthwestern UniversityEvanstonIllinoisUSA
| | - Joshua N. Leonard
- Department of Chemical and Biological EngineeringNorthwestern UniversityEvanstonIllinoisUSA
- Center for Synthetic BiologyNorthwestern UniversityEvanstonIllinoisUSA
- Chemistry of Life Processes InstituteNorthwestern UniversityEvanstonIllinoisUSA
- Interdisciplinary Biological Sciences Training ProgramNorthwestern UniversityEvanstonIllinoisUSA
- Robert H. Lurie Comprehensive Cancer CenterNorthwestern UniversityEvanstonIllinoisUSA
| |
Collapse
|
7
|
Smack C, Johnson B, Nyalwidhe JO, Semmes OJ, Yang L. Small extracellular vesicles: Roles and clinical application in prostate cancer. Adv Cancer Res 2024; 161:119-190. [PMID: 39032949 DOI: 10.1016/bs.acr.2024.05.008] [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: 07/23/2024]
Abstract
Prostate cancer is a significant health problem in the United States. It is remarkably heterogenous, ranging from slow growing disease amenable to active surveillance to highly aggressive forms requiring active treatments. Therefore, being able to precisely determine the nature of disease and appropriately match patients to available and/or novel therapeutics is crucial to improve patients' overall outcome and quality of life. Recently small extracellular vesicles (sEVs), a subset of nanoscale membranous vesicles secreted by various cells, have emerged as important analytes for liquid biopsy and promising vehicles for drug delivery. sEVs contain various biomolecules such as genetic material, proteins, and lipids that recapitulate the characteristics and state of their donor cells. The application of existing and newly developed technologies has resulted in an increased depth of knowledge about biophysical structures, biogenesis, and functions of sEVs. In prostate cancer patients, tumor-derived sEVs can be isolated from biofluids, commonly urine and blood. They mediate intercellular signaling within the tumor microenvironment and distal organ-specific sites, supporting cancer initiation, progression, and metastasis. A mounting body of evidence suggests that sEV components can be potent biomarkers for prostate cancer diagnosis, prognosis, and prediction of disease progression and treatment response. Due to enhanced circulation stability and bio-barrier permeability, sEVs can be also used as effective drug delivery carriers to improve the efficacy and specificity of anti-tumor therapies. This review discusses recent studies on sEVs in prostate cancer and is focused on their role as biomarkers and drug delivery vehicles in the clinical management of prostate cancer.
Collapse
Affiliation(s)
- Caleb Smack
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States; Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Benjamin Johnson
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States; Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Julius O Nyalwidhe
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States; Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - O John Semmes
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States; Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Lifang Yang
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States; Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States.
| |
Collapse
|
8
|
Sharma A, Yadav A, Nandy A, Ghatak S. Insight into the Functional Dynamics and Challenges of Exosomes in Pharmaceutical Innovation and Precision Medicine. Pharmaceutics 2024; 16:709. [PMID: 38931833 PMCID: PMC11206934 DOI: 10.3390/pharmaceutics16060709] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
Of all the numerous nanosized extracellular vesicles released by a cell, the endosomal-originated exosomes are increasingly recognized as potential therapeutics, owing to their inherent stability, low immunogenicity, and targeted delivery capabilities. This review critically evaluates the transformative potential of exosome-based modalities across pharmaceutical and precision medicine landscapes. Because of their precise targeted biomolecular cargo delivery, exosomes are posited as ideal candidates in drug delivery, enhancing regenerative medicine strategies, and advancing diagnostic technologies. Despite the significant market growth projections of exosome therapy, its utilization is encumbered by substantial scientific and regulatory challenges. These include the lack of universally accepted protocols for exosome isolation and the complexities associated with navigating the regulatory environment, particularly the guidelines set forth by the U.S. Food and Drug Administration (FDA). This review presents a comprehensive overview of current research trajectories aimed at addressing these impediments and discusses prospective advancements that could substantiate the clinical translation of exosomal therapies. By providing a comprehensive analysis of both the capabilities and hurdles inherent to exosome therapeutic applications, this article aims to inform and direct future research paradigms, thereby fostering the integration of exosomal systems into mainstream clinical practice.
Collapse
Affiliation(s)
| | | | | | - Subhadip Ghatak
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; (A.S.); (A.Y.); (A.N.)
| |
Collapse
|
9
|
Mitrut RE, Stranford DM, DiBiase BN, Chan JM, Bailey MD, Luo M, Harper CS, Meade TJ, Wang M, Leonard JN. HaloTag display enables quantitative single-particle characterization and functionalization of engineered extracellular vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.25.559433. [PMID: 37808729 PMCID: PMC10557717 DOI: 10.1101/2023.09.25.559433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Extracellular vesicles (EVs) play key roles in diverse biological processes, transport biomolecules between cells, and have been engineered for therapeutic applications. A useful EV bioengineering strategy is to express engineered proteins on the EV surface to confer targeting, bioactivity, and other properties. Measuring how incorporation varies across a population of EVs is important for characterizing such materials and understanding their function, yet it remains challenging to quantitatively characterize the absolute number of engineered proteins incorporated at single-EV resolution. To address these needs, we developed a HaloTag-based characterization platform in which dyes or other synthetic species can be covalently and stoichiometrically attached to engineered proteins on the EV surface. To evaluate this system, we employed several orthogonal quantification methods, including flow cytometry and fluorescence microscopy, and found that HaloTag-mediated quantification is generally robust across EV analysis methods. We compared HaloTag-labeling to antibody-labeling of EVs using single vesicle flow cytometry, enabling us to measure the substantial degree to which antibody labeling can underestimate proteins present on an EV. Finally, we demonstrate the use of HaloTag to compare between protein designs for EV bioengineering. Overall, the HaloTag system is a useful EV characterization tool which complements and expands existing methods.
Collapse
|
10
|
Shiju TM, Yuan A. Extracellular vesicle biomarkers in ocular fluids associated with ophthalmic diseases. Exp Eye Res 2024; 241:109831. [PMID: 38401855 DOI: 10.1016/j.exer.2024.109831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/06/2024] [Accepted: 02/13/2024] [Indexed: 02/26/2024]
Abstract
Extracellular vesicles (EVs) are released as highly stable lipid bilayer particles carrying proteins, lipids, glycans and miRNAs. The contents of EVs vary based on the cellular origin, biogenesis route and the functional state of the cell suggesting certain diseased conditions. A growing body of evidence show that EVs carry important molecules implicated in the development and progression of ophthalmic diseases. EVs associated with ophthalmic diseases are mainly carried by one of the three ocular biofluids which include tears, aqueous humor and vitreous humor. This review summarizes the list of EV derived biomarkers identified thus far in ocular fluids for ophthalmic disease diagnosis. Further, the methods used for sample collection, sample volume and the sample numbers used in these studies have been highlighted. Emphasis has been given to describe the EV isolation and the characterization methods used, EV size profiled and the EV concentrations analyzed by these studies, thus providing a roadmap for future EV biomarker studies in ocular fluids.
Collapse
Affiliation(s)
| | - Alex Yuan
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
11
|
Li Z, Guo K, Gao Z, Chen J, Ye Z, Cao M, Wang SE, Yin Y, Zhong W. Colocalization of protein and microRNA markers reveals unique extracellular vesicle subpopulations for early cancer detection. SCIENCE ADVANCES 2024; 10:eadh8689. [PMID: 38416840 PMCID: PMC10901469 DOI: 10.1126/sciadv.adh8689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 01/25/2024] [Indexed: 03/01/2024]
Abstract
Extracellular vesicles (EVs) play important roles in cell-cell communication but are highly heterogeneous, and each vesicle has dimensions smaller than 200 nm with very limited amounts of cargos encapsulated. The technique of NanOstirBar (NOB)-EnabLed Single Particle Analysis (NOBEL-SPA) reported in the present work permits rapid inspection of single EV with high confidence by confocal fluorescence microscopy, thus enables colocalization assessment for selected protein and microRNA (miRNA) markers in the EVs produced by various cell lines, or present in clinical sera samples. EV subpopulations marked by the colocalization of unique protein and miRNA combinations were discovered to be able to detect early-stage (stage I or II) breast cancer (BC). NOBEL-SPA can be adapted to analyze other types of cargo molecules or other small submicron biological particles. Study of the sorting of specific cargos to heterogeneous vesicles under different physiological conditions can help discover distinct vesicle subpopulations valuable in clinical examination and therapeutics development and gain better understanding of their biogenesis.
Collapse
Affiliation(s)
- Zongbo Li
- Department of Chemistry, University of California-Riverside, Riverside, CA 92521, USA
| | - Kaizhu Guo
- Department of Chemistry, University of California-Riverside, Riverside, CA 92521, USA
| | - Ziting Gao
- Department of Chemistry, University of California-Riverside, Riverside, CA 92521, USA
| | - Junyi Chen
- Environmental Toxicology Graduate Program, University of California-Riverside, Riverside, CA 92521, USA
| | - Zuyang Ye
- Department of Chemistry, University of California-Riverside, Riverside, CA 92521, USA
| | - Minghui Cao
- Department of Pathology, University of California–San Diego, La Jolla, CA 92093, USA
| | - Shizhen Emily Wang
- Department of Pathology, University of California–San Diego, La Jolla, CA 92093, USA
| | - Yadong Yin
- Department of Chemistry, University of California-Riverside, Riverside, CA 92521, USA
| | - Wenwan Zhong
- Department of Chemistry, University of California-Riverside, Riverside, CA 92521, USA
- Environmental Toxicology Graduate Program, University of California-Riverside, Riverside, CA 92521, USA
| |
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: 527] [Impact Index Per Article: 527.0] [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
|
Raju G, Nayak S, Acharya N, Sunder M, Kistenev Y, Mazumder N. Exploring the future of regenerative medicine: Unveiling the potential of optical microscopy for structural and functional imaging of stem cells. JOURNAL OF BIOPHOTONICS 2024; 17:e202300360. [PMID: 38168892 DOI: 10.1002/jbio.202300360] [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: 09/04/2023] [Revised: 10/18/2023] [Accepted: 12/03/2023] [Indexed: 01/05/2024]
Abstract
Regenerative medicine, which utilizes stem cells for tissue and organ repair, holds immense promise in healthcare. A comprehensive understanding of stem cell characteristics is crucial to unlock their potential. This study explores the pivotal role of optical microscopy in advancing regenerative medicine as a potent tool for stem cell research. Advanced optical microscopy techniques enable an in-depth examination of stem cell behavior, morphology, and functionality. The review encompasses current optical microscopy, elucidating its capabilities and constraints in stem cell imaging, while also shedding light on emerging technologies for improved stem cell visualization. Optical microscopy, complemented by techniques like fluorescence and multiphoton imaging, enhances our comprehension of stem cell dynamics. The introduction of label-free imaging facilitates noninvasive, real-time stem cell monitoring without external dyes or markers. By pushing the boundaries of optical microscopy, researchers reveal the intricate cellular mechanisms underpinning regenerative processes, thereby advancing more effective therapeutic strategies. The current study not only outlines the future of regenerative medicine but also underscores the pivotal role of optical microscopy in both structural and functional stem cell imaging.
Collapse
Affiliation(s)
- Gagan Raju
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Smitha Nayak
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Neha Acharya
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Mridula Sunder
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Yury Kistenev
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, Tomsk, Russia
| | - Nirmal Mazumder
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
14
|
Zhang F, Zhang L, Yu H. Potential Druggability of Mesenchymal Stem/Stromal Cell-derived Exosomes. Curr Stem Cell Res Ther 2024; 19:1195-1209. [PMID: 38523514 DOI: 10.2174/011574888x311270240319084835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 03/26/2024]
Abstract
Exosomes secreted by mesenchymal stem/stromal cells (MSC-Exos) are advantageous candidate sources for novel acellular therapy. Despite the current standards of good manufacturing practice (GMP), the deficiency of suitable quality-control methods and the difficulties in large-scale preparation largely restrict the development of therapeutic products and their clinical applications worldwide. Herein, we mainly focus on three dominating issues commonly encountered in exosomal GMP, including issues upstream of the cell culture process, downstream of the purification process, exosomes quality control, and the drug properties of exosomes and their druggability from a corporate perspective. Collectively, in this review article, we put forward the issues of preparing clinical exosome drugs for the treatment of diverse diseases and provide new references for the clinical application of GMP-grade MSC-Exos.
Collapse
Affiliation(s)
- Fan Zhang
- Faculty of Life Sciences and Medicine, Kunming University of Science and Technology, Kunming, 650500, China
| | - Leisheng Zhang
- Science and Technology Innovation Center, The Fourth People's Hospital of Jinan (The Third Affiliated Hospital of Shandong First Medical University), Jinan, 250031, China
- National Health Commission (NHC) Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Hao Yu
- The Postdoctoral Research Station, School of Medicine, Nankai University, Tianjin, 300071, China
| |
Collapse
|
15
|
Lai JJ, Hill JJ, Huang CY, Lee GC, Mai KW, Shen MY, Wang SK. Unveiling the Complex World of Extracellular Vesicles: Novel Characterization Techniques and Manufacturing Considerations. Chonnam Med J 2024; 60:1-12. [PMID: 38304124 PMCID: PMC10828078 DOI: 10.4068/cmj.2024.60.1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/03/2024] Open
Abstract
Extracellular vesicles (EVs) function as potent mediators of intercellular communication for many in vivo processes, contributing to both health and disease related conditions. Given their biological origins and diverse functionality from correspondingly unique "cargo" compositions, both endogenous and modified EVs are garnering attention as promising therapeutic modalities and vehicles for targeted therapeutic delivery applications. Their diversity in composition, however, has revealed a significant need for more comprehensive analytical-based characterization methods, and manufacturing processes that are consistent and scalable. In this review, we explore the dynamic landscape of EV research and development efforts, ranging from novel isolation approaches, to their analytical assessment through novel characterization techniques, and to their production by industrial-scale manufacturing process considerations. Expanding the horizon of these topics to EVs for in-human applications, we underscore the need for stringent development and adherence to Good Manufacturing Practice (GMP) guidelines. Wherein, the intricate interplay of raw materials, production in bioreactors, and isolation practices, along with analytical assessments compliant with the Minimal Information for Studies of Extracellular Vesicles (MISEV) guidelines, in conjunction with reference standard materials, collectively pave the way for standardized and consistent GMP production processes.
Collapse
Affiliation(s)
- James J. Lai
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - John J. Hill
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
- BioProcess Technology Group, BDO, Boston, MA, USA
| | - Casey Y. Huang
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Gino C. Lee
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Karol W. Mai
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Maggie Y. Shen
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Simon K. Wang
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
16
|
Yarana C, Maneechote C, Khuanjing T, Ongnok B, Prathumsap N, Thanasrisuk S, Pattanapanyasat K, Chattipakorn SC, Chattipakorn N. Potential roles of 4HNE-adducted protein in serum extracellular vesicles as an early indicator of oxidative response against doxorubicin-induced cardiomyopathy in rats. Curr Res Toxicol 2023; 5:100134. [PMID: 37964944 PMCID: PMC10641738 DOI: 10.1016/j.crtox.2023.100134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/18/2023] [Accepted: 10/31/2023] [Indexed: 11/16/2023] Open
Abstract
Late-onset cardiomyopathy is becoming more common among cancer survivors, particularly those who received doxorubicin (DOXO) treatment. However, few clinically available cardiac biomarkers can predict an unfavorable cardiac outcome before cell death. Extracellular vesicles (EVs) are emerging as biomarkers for cardiovascular diseases and others. This study aimed to measure dynamic 4-hydroxynonenal (4HNE)-adducted protein levels in rats treated chronically with DOXO and examine their link with oxidative stress, antioxidant gene expression in cardiac tissues, and cardiac function. Twenty-two male Wistar rats were randomly assigned to receive intraperitoneal injection of normal saline (n = 8) or DOXO (3 mg/kg, 6 doses, n = 14). Before and after therapy, serum EVs and N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels were determined. Tunable resistive pulse sensing was used to measure EV size and concentration. ELISA was used to assess 4HNE-adducted protein in EVs and cardiac tissues. Differential-display reverse transcription-PCR was used to quantitate cardiac Cat and Gpx1 gene expression. Potential correlations between 4HNE-adducted protein levels in EVs, cardiac oxidative stress, antioxidant gene expression, and cardiac function were determined. DOXO-treated rats showed more serum EV 4HNE-adducted protein than NSS-treated rats at day 9 and later endpoints, whereas NT-proBNP levels were not different between groups. Moreover, on day 9, surviving rats' EVs had higher levels of 4HNE-adducted protein, and these correlated positively with concentrations of heart tissue 4HNE adduction and copy numbers of Cat and Gpx1, while at endpoint correlated negatively with cardiac functions. Therefore, 4HNE-adducted protein in serum EVs could be an early, minimally invasive biomarker of the oxidative response and cardiac function in DOXO-induced cardiomyopathy.
Collapse
Affiliation(s)
- Chontida Yarana
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, 999 Phuttamonthon 4 Road, Salaya, Nakhon Pathom 73170, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Thawatchai Khuanjing
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Benjamin Ongnok
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nanthip Prathumsap
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sirasa Thanasrisuk
- Faculty of Medical Technology, Mahidol University, 999 Phuttamonthon 4 Road, Salaya, Nakhon Pathom 73170, Thailand
| | - Kovit Pattanapanyasat
- Center of Excellence for Microparticle and Exosome in Diseases, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Siriporn C. Chattipakorn
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
17
|
Zhang J, Wu J, Wang G, He L, Zheng Z, Wu M, Zhang Y. Extracellular Vesicles: Techniques and Biomedical Applications Related to Single Vesicle Analysis. ACS NANO 2023; 17:17668-17698. [PMID: 37695614 DOI: 10.1021/acsnano.3c03172] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Extracellular vesicles (EVs) are extensively dispersed lipid bilayer membrane vesicles involved in the delivery and transportation of molecular payloads to certain cell types to facilitate intercellular interactions. Their significant roles in physiological and pathological processes make EVs outstanding biomarkers for disease diagnosis and treatment monitoring as well as ideal candidates for drug delivery. Nevertheless, differences in the biogenesis processes among EV subpopulations have led to a diversity of biophysical characteristics and molecular cargos. Additionally, the prevalent heterogeneity of EVs has been found to substantially hamper the sensitivity and accuracy of disease diagnosis and therapeutic monitoring, thus impeding the advancement of clinical applications. In recent years, the evolution of single EV (SEV) analysis has enabled an in-depth comprehension of the physical properties, molecular composition, and biological roles of EVs at the individual vesicle level. This review examines the sample acquisition tactics prior to SEV analysis, i.e., EV isolation techniques, and outlines the current state-of-the-art label-free and label-based technologies for SEV identification. Furthermore, the challenges and prospects of biomedical applications based on SEV analysis are systematically discussed.
Collapse
Affiliation(s)
- Jie Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Jiacheng Wu
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Guanzhao Wang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Luxuan He
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Ziwei Zheng
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Minhao Wu
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, P. R. China
| | - Yuanqing Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| |
Collapse
|
18
|
Cheng T. Single-molecule localization microscopy based on denoising, interpolation and local maxima. Microscopy (Oxf) 2023; 72:336-342. [PMID: 36412750 DOI: 10.1093/jmicro/dfac065] [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: 09/05/2022] [Revised: 11/05/2022] [Accepted: 11/21/2022] [Indexed: 08/05/2023] Open
Abstract
A single fluorescent molecule is highly likely to be located at the center pixel position of a raw image diffused spot in an ideal situation. Even if the molecule and the center pixel position do not completely overlap, they are very close. A single-molecule localization method based on denoising, interpolation and local maxima (DIL) is proposed. The low-resolution raw image is denoised and interpolated, and a new image with a pixel size equal to that of the super-resolution image is attained. The local maxima of the new image are extracted. With this method, it is found that the local maxima positions can be regarded as the fluorescent molecule positions. Simulation results demonstrate that the DIL single-molecule localization accuracy reaches ∼18 nm when the Gaussian noise variance is equal to 0.01. Experimental results demonstrate that the DIL localization methodology is comparable to the Gaussian fitting algorithm and is faster.
Collapse
Affiliation(s)
- Tao Cheng
- School of Mechanical and Automotive Engineering, Guangxi University of Science and Technology, No. 268 Avenue Donghuan, Chengzhong District, Liuzhou, Guangxi 545006, P. R. China
| |
Collapse
|
19
|
Saftics A, Abuelreich S, Romano E, Ghaeli I, Jiang N, Spanos M, Lennon KM, Singh G, Das S, Van Keuren‐Jensen K, Jovanovic‐Talisman T. Single Extracellular VEsicle Nanoscopy. J Extracell Vesicles 2023; 12:e12346. [PMID: 37422692 PMCID: PMC10329735 DOI: 10.1002/jev2.12346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 05/26/2023] [Accepted: 06/23/2023] [Indexed: 07/10/2023] Open
Abstract
Extracellular vesicles (EVs) and their cargo constitute novel biomarkers. EV subpopulations have been defined not only by abundant tetraspanins (e.g., CD9, CD63 and CD81) but also by specific markers derived from their source cells. However, it remains a challenge to robustly isolate and characterize EV subpopulations. Here, we combined affinity isolation with super-resolution imaging to comprehensively assess EV subpopulations from human plasma. Our Single Extracellular VEsicle Nanoscopy (SEVEN) assay successfully quantified the number of affinity-isolated EVs, their size, shape, molecular tetraspanin content, and heterogeneity. The number of detected tetraspanin-enriched EVs positively correlated with sample dilution in a 64-fold range (for SEC-enriched plasma) and a 50-fold range (for crude plasma). Importantly, SEVEN robustly detected EVs from as little as ∼0.1 μL of crude plasma. We further characterized the size, shape and molecular tetraspanin content (with corresponding heterogeneities) for CD9-, CD63- and CD81-enriched EV subpopulations. Finally, we assessed EVs from the plasma of four pancreatic ductal adenocarcinoma patients with resectable disease. Compared to healthy plasma, CD9-enriched EVs from patients were smaller while IGF1R-enriched EVs from patients were larger, rounder and contained more tetraspanin molecules, suggestive of a unique pancreatic cancer-enriched EV subpopulation. This study provides the method validation and demonstrates that SEVEN could be advanced into a platform for characterizing both disease-associated and organ-associated EV subpopulations.
Collapse
Affiliation(s)
- Andras Saftics
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Sarah Abuelreich
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Eugenia Romano
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Ima Ghaeli
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Nan Jiang
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Michail Spanos
- Cardiology Division and Corrigan Minehan Heart CenterMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Kathleen M. Lennon
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Gagandeep Singh
- Department of SurgeryCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Saumya Das
- Cardiology Division and Corrigan Minehan Heart CenterMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | | | - Tijana Jovanovic‐Talisman
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| |
Collapse
|
20
|
Lauriola A, Davalli P, Marverti G, Santi S, Caporali A, D'Arca D. Targeting the Interplay of Independent Cellular Pathways and Immunity: A Challenge in Cancer Immunotherapy. Cancers (Basel) 2023; 15:cancers15113009. [PMID: 37296972 DOI: 10.3390/cancers15113009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 05/19/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023] Open
Abstract
Immunotherapy is a cancer treatment that exploits the capacity of the body's immune system to prevent, control, and remove cancer. Immunotherapy has revolutionized cancer treatment and significantly improved patient outcomes for several tumor types. However, most patients have not benefited from such therapies yet. Within the field of cancer immunotherapy, an expansion of the combination strategy that targets independent cellular pathways that can work synergistically is predicted. Here, we review some consequences of tumor cell death and increased immune system engagement in the modulation of oxidative stress and ubiquitin ligase pathways. We also indicate combinations of cancer immunotherapies and immunomodulatory targets. Additionally, we discuss imaging techniques, which are crucial for monitoring tumor responses during treatment and the immunotherapy side effects. Finally, the major outstanding questions are also presented, and directions for future research are described.
Collapse
Affiliation(s)
- Angela Lauriola
- Department of Biotechnology, University of Verona, 37134 Verona, Italy
| | - Pierpaola Davalli
- Department of Biomedical, Metabolic and Neural Sciences, Via G. Campi 287, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Gaetano Marverti
- Department of Biomedical, Metabolic and Neural Sciences, Via G. Campi 287, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Spartaco Santi
- Consiglio Nazionale delle Ricerche (CNR) Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", 40136 Bologna, Italy
- IRCCS, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Andrea Caporali
- BHF Centre for Cardiovascular Science, University of Edinburgh, Scotland EH4 2XU, UK
| | - Domenico D'Arca
- Department of Biomedical, Metabolic and Neural Sciences, Via G. Campi 287, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
21
|
Kim J, Kang MS, Jun SW, Jo HJ, Han DW, Kim CS. A systematic study on the use of multifunctional nanodiamonds for neuritogenesis and super-resolution imaging. Biomater Res 2023; 27:37. [PMID: 37106432 PMCID: PMC10134586 DOI: 10.1186/s40824-023-00384-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Regeneration of defective neurons in central nervous system is a highlighted issue for neurodegenerative disease treatment. Various tissue engineering approaches have focused on neuritogenesis to achieve the regeneration of damaged neuronal cells because damaged neurons often fail to achieve spontaneous restoration of neonatal neurites. Meanwhile, owing to the demand for a better diagnosis, studies of super-resolution imaging techniques in fluorescence microscopy have triggered the technological development to surpass the classical resolution dictated by the optical diffraction limit for precise observations of neuronal behaviors. Herein, the multifunctional nanodiamonds (NDs) as neuritogenesis promoters and super-resolution imaging probes were studied. METHODS To investigate the neuritogenesis-inducing capability of NDs, ND-containing growing medium and differentiation medium were added to the HT-22 hippocampal neuronal cells and incubated for 10 d. In vitro and ex vivo images were visualized through custom-built two-photon microscopy using NDs as imaging probes and the direct stochastic optical reconstruction microscopy (dSTORM) process was performed for the super-resolution reconstruction owing to the photoblinking properties of NDs. Moreover, ex vivo imaging of the mouse brain was performed 24 h after the intravenous injection of NDs. RESULTS NDs were endocytosed by the cells and promoted spontaneous neuritogenesis without any differentiation factors, where NDs exhibited no significant toxicity with their outstanding biocompatibility. The images of ND-endocytosed cells were reconstructed into super-resolution images through dSTORM, thereby addressing the problem of image distortion due to nano-sized particles, including size expansion and the challenge in distinguishing the nearby located particles. Furthermore, the ex vivo images of NDs in mouse brain confirmed that NDs could penetrate the blood-brain barrier (BBB) and retain their photoblinking property for dSTORM application. CONCLUSIONS It was demonstrated that the NDs are capable of dSTORM super-resolution imaging, neuritogenic facilitation, and BBB penetration, suggesting their remarkable potential in biological applications.
Collapse
Affiliation(s)
- Jaeheung Kim
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, 46241, Republic of Korea
| | - Moon Sung Kang
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, 46241, Republic of Korea
| | - Seung Won Jun
- Agency for Defense Development, Ground Technology Research Institute, Daejeon, 34186, Republic of Korea
| | - Hyo Jung Jo
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, 46241, Republic of Korea
| | - Dong-Wook Han
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, 46241, Republic of Korea.
- Bio-IT Fusion Technology Research Institute, Pusan National University, Busan, 46241, Republic of Korea.
| | - Chang-Seok Kim
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, 46241, Republic of Korea.
- Engineering Research Center for Color-Modulated Extra-Sensory Perception Technology, Pusan National University, Busan, 46241, Republic of Korea.
| |
Collapse
|
22
|
Li Z, Guo K, Gao Z, Chen J, Ye Z, Wang SE, Yin Y, Zhong W. Colocalization of Protein and microRNA Markers Reveals Unique Extracellular Vesicle Sub-Populations for Early Cancer Detection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.536958. [PMID: 37131582 PMCID: PMC10153150 DOI: 10.1101/2023.04.17.536958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Extracellular vesicles (EVs) play important roles in cell-cell communication but they are highly heterogeneous, and each vesicle has dimensions smaller than 200 nm thus encapsulates very limited amounts of cargos. We report the technique of NanOstirBar (NOB)-EnabLed Single Particle Analysis (NOBEL-SPA) that utilizes NOBs, which are superparamagnetic nanorods easily handled by a magnet or a rotating magnetic field, to act as isolated "islands" for EV immobilization and cargo confinement. NOBEL-SPA permits rapid inspection of single EV with high confidence by confocal fluorescence microscopy, and can assess the colocalization of selected protein/microRNA (miRNA) pairs in the EVs produced by various cell lines or present in clinical sera samples. Specific EV sub-populations marked by the colocalization of unique protein and miRNA combinations have been revealed by the present work, which can differentiate the EVs by their cells or origin, as well as to detect early-stage breast cancer (BC). We believe NOBEL-SPA can be expanded to analyze the co-localization of other types of cargo molecules, and will be a powerful tool to study EV cargo loading and functions under different physiological conditions, and help discover distinct EV subgroups valuable in clinical examination and therapeutics development.
Collapse
|
23
|
Pucci M, Moschetti M, Urzì O, Loria M, Conigliaro A, Di Bella MA, Crescitelli R, Olofsson Bagge R, Gallo A, Santos MF, Puglisi C, Forte S, Lorico A, Alessandro R, Fontana S. Colorectal cancer-derived small extracellular vesicles induce TGFβ1-mediated epithelial to mesenchymal transition of hepatocytes. Cancer Cell Int 2023; 23:77. [PMID: 37072829 PMCID: PMC10114452 DOI: 10.1186/s12935-023-02916-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 03/31/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Metastatic disease is the major cause of cancer-related deaths. Increasing evidence shows that primary tumor cells can promote metastasis by preparing the local microenvironment of distant organs, inducing the formation of the so-called "pre-metastatic niche". In recent years, several studies have highlighted that among the tumor-derived molecular components active in pre-metastatic niche formation, small extracellular vesicles (sEVs) play a crucial role. Regarding liver metastasis, the ability of tumor-derived sEVs to affect the activities of non-parenchymal cells such as Kupffer cells and hepatic stellate cells is well described, while the effects on hepatocytes, the most conspicuous and functionally relevant hepatic cellular component, remain unknown. METHODS sEVs isolated from SW480 and SW620 CRC cells and from clinical samples of CRC patients and healthy subjects were used to treat human healthy hepatocytes (THLE-2 cells). RT-qPCR, Western blot and confocal microscopy were applied to investigate the effects of this treatment. RESULTS Our study shows for the first time that TGFβ1-carrying CRC_sEVs impair the morphological and functional properties of healthy human hepatocytes by triggering their TGFβ1/SMAD-dependent EMT. These abilities of CRC_sEVs were further confirmed by evaluating the effects elicited on hepatocytes by sEVs isolated from plasma and biopsies from CRC patients. CONCLUSIONS Since it is known that EMT of hepatocytes leads to the formation of a fibrotic environment, a well-known driver of metastasis, these results suggest that CRC_sEV-educated hepatocytes could have an active and until now neglected role during liver metastasis formation.
Collapse
Affiliation(s)
- Marzia Pucci
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Marta Moschetti
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Ornella Urzì
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Marco Loria
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Alice Conigliaro
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Maria Antonietta Di Bella
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Rossella Crescitelli
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Roger Olofsson Bagge
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Alessia Gallo
- Department of Research, IRCCS ISMETT, Palermo, Italy
| | - Mark F Santos
- Touro University College of Medicine, Henderson, NV, USA
| | | | | | - Aurelio Lorico
- Touro University College of Medicine, Henderson, NV, USA
- IOM Ricerca, Viagrande, Catania, Italy
| | - Riccardo Alessandro
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Palermo, Italy
| | - Simona Fontana
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy.
| |
Collapse
|
24
|
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
|
25
|
Spitzberg JD, Ferguson S, Yang KS, Peterson HM, Carlson JCT, Weissleder R. Multiplexed analysis of EV reveals specific biomarker composition with diagnostic impact. Nat Commun 2023; 14:1239. [PMID: 36870999 PMCID: PMC9985597 DOI: 10.1038/s41467-023-36932-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Exosomes and extracellular vesicles (EV) are increasingly being explored as circulating biomarkers, but their heterogenous composition will likely mandate the development of multiplexed EV technologies. Iteratively multiplexed analyses of near single EVs have been challenging to implement beyond a few colors during spectral sensing. Here we developed a multiplexed analysis of EV technique (MASEV) to interrogate thousands of individual EVs during 5 cycles of multi-channel fluorescence staining for 15 EV biomarkers. Contrary to the common belief, we show that: several markers proposed to be ubiquitous are less prevalent than believed; multiple biomarkers concur in single vesicles but only in small fractions; affinity purification can lead to loss of rare EV subtypes; and deep profiling allows detailed analysis of EV, potentially improving the diagnostic content. These findings establish the potential of MASEV for uncovering fundamental EV biology and heterogeneity and increasing diagnostic specificity.
Collapse
Affiliation(s)
- Joshua D Spitzberg
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA, 02114, USA
| | - Scott Ferguson
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA, 02114, USA
| | - Katherine S Yang
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA, 02114, USA
| | - Hannah M Peterson
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA, 02114, USA
| | - Jonathan C T Carlson
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA, 02114, USA. .,Cancer Center, Massachusetts General Hospital, Boston, MA, 02114, USA.
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA, 02114, USA. .,Cancer Center, Massachusetts General Hospital, Boston, MA, 02114, USA. .,Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA, 02115, USA.
| |
Collapse
|
26
|
Ghanam J, Chetty VK, Zhu X, Liu X, Gelléri M, Barthel L, Reinhardt D, Cremer C, Thakur BK. Single Molecule Localization Microscopy for Studying Small Extracellular Vesicles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205030. [PMID: 36635058 DOI: 10.1002/smll.202205030] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/23/2022] [Indexed: 06/17/2023]
Abstract
Small extracellular vesicles (sEVs) are 30-200 nm nanovesicles enriched with unique cargoes of nucleic acids, lipids, and proteins. sEVs are released by all cell types and have emerged as a critical mediator of cell-to-cell communication. Although many studies have dealt with the role of sEVs in health and disease, the exact mechanism of sEVs biogenesis and uptake remain unexplored due to the lack of suitable imaging technologies. For sEVs functional studies, imaging has long relied on conventional fluorescence microscopy that has only 200-300 nm resolution, thereby generating blurred images. To break this resolution limit, recent developments in super-resolution microscopy techniques, specifically single-molecule localization microscopy (SMLM), expanded the understanding of subcellular details at the few nanometer level. SMLM success relies on the use of appropriate fluorophores with excellent blinking properties. In this review, the basic principle of SMLM is highlighted and the state of the art of SMLM use in sEV biology is summarized. Next, how SMLM techniques implemented for cell imaging can be translated to sEV imaging is discussed by applying different labeling strategies to study sEV biogenesis and their biomolecular interaction with the distant recipient cells.
Collapse
Affiliation(s)
- Jamal Ghanam
- Department of Pediatrics III, University Hospital Essen, 45147, Essen, Germany
| | | | - Xingfu Zhu
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | - Xiaomin Liu
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | - Márton Gelléri
- Institute of Molecular Biology (IMB), 55128, Mainz, Germany
| | - Lennart Barthel
- Department of Neurosurgery and Spine Surgery, Center for Translational Neuro and Behavioral Sciences, University Hospital Essen, 45147, Essen, Germany
- Institute of Medical Psychology and Behavioral Immunobiology, Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, 45147, Essen, Germany
| | - Dirk Reinhardt
- Department of Pediatrics III, University Hospital Essen, 45147, Essen, Germany
| | - Christoph Cremer
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
- Institute of Molecular Biology (IMB), 55128, Mainz, Germany
| | - Basant Kumar Thakur
- Department of Pediatrics III, University Hospital Essen, 45147, Essen, Germany
| |
Collapse
|
27
|
Blommer J, Pitcher T, Mustapic M, Eren E, Yao PJ, Vreones MP, Pucha KA, Dalrymple-Alford J, Shoorangiz R, Meissner WG, Anderson T, Kapogiannis D. Extracellular vesicle biomarkers for cognitive impairment in Parkinson's disease. Brain 2023; 146:195-208. [PMID: 35833836 PMCID: PMC10060702 DOI: 10.1093/brain/awac258] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/24/2022] [Accepted: 06/22/2022] [Indexed: 01/11/2023] Open
Abstract
Besides motor symptoms, many individuals with Parkinson's disease develop cognitive impairment perhaps due to coexisting α-synuclein and Alzheimer's disease pathologies and impaired brain insulin signalling. Discovering biomarkers for cognitive impairment in Parkinson's disease could help clarify the underlying pathogenic processes and improve Parkinson's disease diagnosis and prognosis. This study used plasma samples from 273 participants: 103 Parkinson's disease individuals with normal cognition, 121 Parkinson's disease individuals with cognitive impairment (81 with mild cognitive impairment, 40 with dementia) and 49 age- and sex-matched controls. Plasma extracellular vesicles enriched for neuronal origin were immunocaptured by targeting the L1 cell adhesion molecule, then biomarkers were quantified using immunoassays. α-Synuclein was lower in Parkinson's disease compared to control individuals (P = 0.004) and in cognitively impaired Parkinson's disease individuals compared to Parkinson's disease with normal cognition (P < 0.001) and control (P < 0.001) individuals. Amyloid-β42 did not differ between groups. Phosphorylated tau (T181) was higher in Parkinson's disease than control individuals (P = 0.003) and in cognitively impaired compared to cognitively normal Parkinson's disease individuals (P < 0.001) and controls (P < 0.001). Total tau was not different between groups. Tyrosine-phosphorylated insulin receptor substrate-1 was lower in Parkinson's disease compared to control individuals (P = 0.03) and in cognitively impaired compared to cognitively normal Parkinson's disease individuals (P = 0.02) and controls (P = 0.01), and also decreased with increasing motor symptom severity (P = 0.005); serine312-phosphorylated insulin receptor substrate-1 was not different between groups. Mechanistic target of rapamycin was not different between groups, whereas phosphorylated mechanistic target of rapamycin trended lower in cognitively impaired compared to cognitively normal Parkinson's disease individuals (P = 0.05). The ratio of α-synuclein to phosphorylated tau181 was lower in Parkinson's disease compared to controls (P = 0.001), in cognitively impaired compared to cognitively normal Parkinson's disease individuals (P < 0.001) and decreased with increasing motor symptom severity (P < 0.001). The ratio of insulin receptor substrate-1 phosphorylated serine312 to insulin receptor substrate-1 phosphorylated tyrosine was higher in Parkinson's disease compared to control individuals (P = 0.01), in cognitively impaired compared to cognitively normal Parkinson's disease individuals (P = 0.02) and increased with increasing motor symptom severity (P = 0.003). α-Synuclein, phosphorylated tau181 and insulin receptor substrate-1 phosphorylated tyrosine contributed in diagnostic classification between groups. These findings suggest that both α-synuclein and tau pathologies and impaired insulin signalling underlie Parkinson's disease with cognitive impairment. Plasma neuronal extracellular vesicles biomarkers may inform cognitive prognosis in Parkinson's disease.
Collapse
Affiliation(s)
- Joseph Blommer
- National Institute on Aging, Intramural Research Program, Laboratory of Clinical Investigation, Baltimore, MD 21224, USA
| | - Toni Pitcher
- New Zealand Brain Research Institute, Christchurch 8011, New Zealand
- Department of Medicine, University of Otago, Christchurch 8011, New Zealand
| | - Maja Mustapic
- National Institute on Aging, Intramural Research Program, Laboratory of Clinical Investigation, Baltimore, MD 21224, USA
| | - Erden Eren
- National Institute on Aging, Intramural Research Program, Laboratory of Clinical Investigation, Baltimore, MD 21224, USA
| | - Pamela J Yao
- National Institute on Aging, Intramural Research Program, Laboratory of Clinical Investigation, Baltimore, MD 21224, USA
| | - Michael P Vreones
- National Institute on Aging, Intramural Research Program, Laboratory of Clinical Investigation, Baltimore, MD 21224, USA
| | - Krishna A Pucha
- National Institute on Aging, Intramural Research Program, Laboratory of Clinical Investigation, Baltimore, MD 21224, USA
| | - John Dalrymple-Alford
- New Zealand Brain Research Institute, Christchurch 8011, New Zealand
- School of Psychology, Speech and Hearing, University of Canterbury, Christchurch 8041, New Zealand
| | - Reza Shoorangiz
- New Zealand Brain Research Institute, Christchurch 8011, New Zealand
| | - Wassilios G Meissner
- New Zealand Brain Research Institute, Christchurch 8011, New Zealand
- University of Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
- Service de Neurologie—Maladies Neurodégénératives, CHU Bordeaux, F-33000 Bordeaux, France
| | - Tim Anderson
- New Zealand Brain Research Institute, Christchurch 8011, New Zealand
- Department of Medicine, University of Otago, Christchurch 8011, New Zealand
| | - Dimitrios Kapogiannis
- National Institute on Aging, Intramural Research Program, Laboratory of Clinical Investigation, Baltimore, MD 21224, USA
| |
Collapse
|
28
|
De Sousa KP, Rossi I, Abdullahi M, Ramirez MI, Stratton D, Inal JM. Isolation and characterization of extracellular vesicles and future directions in diagnosis and therapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1835. [PMID: 35898167 PMCID: PMC10078256 DOI: 10.1002/wnan.1835] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/23/2022] [Accepted: 06/30/2022] [Indexed: 01/31/2023]
Abstract
Extracellular vesicles (EVs) are a unique and heterogeneous class of lipid bilayer nanoparticles secreted by most cells. EVs are regarded as important mediators of intercellular communication in both prokaryotic and eukaryotic cells due to their ability to transfer proteins, lipids and nucleic acids to recipient cells. In addition to their physiological role, EVs are recognized as modulators in pathological processes such as cancer, infectious diseases, and neurodegenerative disorders, providing new potential targets for diagnosis and therapeutic intervention. For a complete understanding of EVs as a universal cellular biological system and its translational applications, optimal techniques for their isolation and characterization are required. Here, we review recent progress in those techniques, from isolation methods to characterization techniques. With interest in therapeutic applications of EVs growing, we address fundamental points of EV-related cell biology, such as cellular uptake mechanisms and their biodistribution in tissues as well as challenges to their application as drug carriers or biomarkers for less invasive diagnosis or as immunogens. This article is categorized under: Diagnostic Tools > Biosensing Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease.
Collapse
Affiliation(s)
- Karina P. De Sousa
- Bioscience Research Group, School of Life and Medical SciencesUniversity of HertfordshireHertfordshireUK
| | - Izadora Rossi
- School of Human SciencesLondon Metropolitan UniversityLondonUK
- Federal University of ParanáCuritibaBrazil
| | | | - Marcel Ivan Ramirez
- Federal University of ParanáCuritibaBrazil
- Carlos Chagas Institute (ICC)CuritibaBrazil
| | - Dan Stratton
- Open UniversityThe School of Life, Health and Chemical SciencesMilton KeynesUK
| | - Jameel Malhador Inal
- Bioscience Research Group, School of Life and Medical SciencesUniversity of HertfordshireHertfordshireUK
- School of Human SciencesLondon Metropolitan UniversityLondonUK
| |
Collapse
|
29
|
Single-cell extracellular vesicle analysis by microfluidics and beyond. Trends Analyt Chem 2023. [DOI: 10.1016/j.trac.2023.116930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
30
|
Surface protein profiling of prostate-derived extracellular vesicles by mass spectrometry and proximity assays. Commun Biol 2022; 5:1402. [PMID: 36550367 PMCID: PMC9780212 DOI: 10.1038/s42003-022-04349-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Extracellular vesicles (EVs) are mediators of intercellular communication and a promising class of biomarkers. Surface proteins of EVs play decisive roles in establishing a connection with recipient cells, and they are putative targets for diagnostic assays. Analysis of the surface proteins can thus both illuminate the biological functions of EVs and help identify potential biomarkers. We developed a strategy combining high-resolution mass spectrometry (HRMS) and proximity ligation assays (PLA) to first identify and then validate surface proteins discovered on EVs. We applied our workflow to investigate surface proteins of small EVs found in seminal fluid (SF-sEV). We identified 1,014 surface proteins and verified the presence of a subset of these on the surface of SF-sEVs. Our work demonstrates a general strategy for deep analysis of EVs' surface proteins across patients and pathological conditions, proceeding from unbiased screening by HRMS to ultra-sensitive targeted analyses via PLA.
Collapse
|
31
|
Mohamad Zamani NS, Wan Zaki WMD, Abd Hamid Z, Baseri Huddin A. Future stem cell analysis: progress and challenges towards state-of-the art approaches in automated cells analysis. PeerJ 2022; 10:e14513. [PMID: 36573241 PMCID: PMC9789697 DOI: 10.7717/peerj.14513] [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: 09/22/2022] [Accepted: 11/14/2022] [Indexed: 12/24/2022] Open
Abstract
Background and Aims A microscopic image has been used in cell analysis for cell type identification and classification, cell counting and cell size measurement. Most previous research works are tedious, including detailed understanding and time-consuming. The scientists and researchers are seeking modern and automatic cell analysis approaches in line with the current in-demand technology. Objectives This article provides a brief overview of a general cell and specific stem cell analysis approaches from the history of cell discovery up to the state-of-the-art approaches. Methodology A content description of the literature study has been surveyed from specific manuscript databases using three review methods: manuscript identification, screening, and inclusion. This review methodology is based on Prism guidelines in searching for originality and novelty in studies concerning cell analysis. Results By analysing generic cell and specific stem cell analysis approaches, current technology offers tremendous potential in assisting medical experts in performing cell analysis using a method that is less laborious, cost-effective, and reduces error rates. Conclusion This review uncovers potential research gaps concerning generic cell and specific stem cell analysis. Thus, it could be a reference for developing automated cells analysis approaches using current technology such as artificial intelligence and deep learning.
Collapse
Affiliation(s)
- Nurul Syahira Mohamad Zamani
- Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, Department of Electrical, Electronic and Systems Engineering, UKM Bangi, Selangor, Malaysia
| | - Wan Mimi Diyana Wan Zaki
- Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, Department of Electrical, Electronic and Systems Engineering, UKM Bangi, Selangor, Malaysia
| | - Zariyantey Abd Hamid
- Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Biomedical Science Programme and Centre for Diagnostic, Therapeutic and Investigative Science, Kuala Lumpur, W. P. Kuala Lumpur, Malaysia
| | - Aqilah Baseri Huddin
- Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, Department of Electrical, Electronic and Systems Engineering, UKM Bangi, Selangor, Malaysia
| |
Collapse
|
32
|
Matsuzaka Y, Yashiro R. Advances in Purification, Modification, and Application of Extracellular Vesicles for Novel Clinical Treatments. MEMBRANES 2022; 12:membranes12121244. [PMID: 36557150 PMCID: PMC9787595 DOI: 10.3390/membranes12121244] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 06/01/2023]
Abstract
Extracellular vesicles (EV) are membrane vesicles surrounded by a lipid bilayer membrane and include microvesicles, apoptotic bodies, exosomes, and exomeres. Exosome-encapsulated microRNAs (miRNAs) released from cancer cells are involved in the proliferation and metastasis of tumor cells via angiogenesis. On the other hand, mesenchymal stem cell (MSC) therapy, which is being employed in regenerative medicine owing to the ability of MSCs to differentiate into various cells, is due to humoral factors, including messenger RNA (mRNA), miRNAs, proteins, and lipids, which are encapsulated in exosomes derived from transplanted cells. New treatments that advocate cell-free therapy using MSC-derived exosomes will significantly improve clinical practice. Therefore, using highly purified exosomes that perform their original functions is desirable. In this review, we summarized advances in the purification, modification, and application of EVs as novel strategies to treat some diseases.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-0031, Japan
| | - Ryu Yashiro
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-0031, Japan
- Department of Infectious Diseases, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-shi, Tokyo 181-0004, Japan
| |
Collapse
|
33
|
Mecocci S, Trabalza-Marinucci M, Cappelli K. Extracellular Vesicles from Animal Milk: Great Potentialities and Critical Issues. Animals (Basel) 2022; 12:ani12233231. [PMID: 36496752 PMCID: PMC9740508 DOI: 10.3390/ani12233231] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 10/25/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
Other than representing the main source of nutrition for newborn mammals, milk delivers a sophisticated signaling system from mother to child that promotes postnatal health. The bioactive components transferred through the milk intake are important for the development of the newborn immune system and include oligosaccharides, lactoferrin, lysozyme, α-La, and immunoglobulins. In the last 15 years, a pivotal role in this mother-to-child exchange has been attributed to extracellular vesicles (EVs). EVs are micro- and nanosized structures enclosed in a phospholipidic double-layer membrane that are produced by all cell types and released in the extracellular environment, reaching both close and distant cells. EVs mediate the intercellular cross-talk from the producing to the receiving cell through the transfer of molecules contained within them such as proteins, antigens, lipids, metabolites, RNAs, and DNA fragments. The complex cargo can induce a wide range of functional modulations in the recipient cell (i.e., anti-inflammatory, immunomodulating, angiogenetic, and pro-regenerative modulations) depending on the type of producing cells and the stimuli that these cells receive. EVs can be recovered from every biological fluid, including blood, urine, bronchoalveolar lavage fluid, saliva, bile, and milk, which is one of the most promising scalable vesicle sources. This review aimed to present the state-of-the-art of animal-milk-derived EV (mEV) studies due to the exponential growth of this field. A focus on the beneficial potentialities for human health and the issues of studying vesicles from milk, particularly for the analytical methodologies applied, is reported.
Collapse
|
34
|
Capra J, Härkönen K, Kyykallio H, Vihinen H, Jokitalo E, Rilla K. Microscopic characterization reveals the diversity of EVs secreted by GFP-HAS3 expressing MCF7 cells. Eur J Cell Biol 2022; 101:151235. [DOI: 10.1016/j.ejcb.2022.151235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 11/27/2022] Open
|
35
|
Yang J, Xu Y. Nanofluidics for sub-single cellular studies: Nascent progress, critical technologies, and future perspectives. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.09.066] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
36
|
Imanbekova M, Suarasan S, Lu Y, Jurchuk S, Wachsmann-Hogiu S. Recent advances in optical label-free characterization of extracellular vesicles. NANOPHOTONICS 2022; 11:2827-2863. [PMID: 35880114 PMCID: PMC9128385 DOI: 10.1515/nanoph-2022-0057] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/16/2022] [Indexed: 05/04/2023]
Abstract
Extracellular vesicles (EVs) are complex biological nanoparticles endogenously secreted by all eukaryotic cells. EVs carry a specific molecular cargo of proteins, lipids, and nucleic acids derived from cells of origin and play a significant role in the physiology and pathology of cells, organs, and organisms. Upon release, they may be found in different body fluids that can be easily accessed via noninvasive methodologies. Due to the unique information encoded in their molecular cargo, they may reflect the state of the parent cell and therefore EVs are recognized as a rich source of biomarkers for early diagnostics involving liquid biopsy. However, body fluids contain a mixture of EVs released by different types of healthy and diseased cells, making the detection of the EVs of interest very challenging. Recent research efforts have been focused on the detection and characterization of diagnostically relevant subpopulations of EVs, with emphasis on label-free methods that simplify sample preparation and are free of interfering signals. Therefore, in this paper, we review the recent progress of the label-free optical methods employed for the detection, counting, and morphological and chemical characterization of EVs. We will first briefly discuss the biology and functions of EVs, and then introduce different optical label-free techniques for rapid, precise, and nondestructive characterization of EVs such as nanoparticle tracking analysis, dynamic light scattering, atomic force microscopy, surface plasmon resonance spectroscopy, Raman spectroscopy, and SERS spectroscopy. In the end, we will discuss their applications in the detection of neurodegenerative diseases and cancer and provide an outlook on the future impact and challenges of these technologies to the field of liquid biopsy via EVs.
Collapse
Affiliation(s)
- Meruyert Imanbekova
- Bioengineering, McGill University Faculty of Engineering, Montreal, QC, Canada
| | - Sorina Suarasan
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, T. Laurian 42, 400271, Cluj-Napoca, Romania
| | - Yao Lu
- Bioengineering, McGill University Faculty of Engineering, 3480 Rue Universite, 1006, Montreal, QC, H3C6W1, Canada
| | - Sarah Jurchuk
- Bioengineering, McGill University Faculty of Engineering, 3480 Rue Universite, Rm#350, Montreal, QC, H3A 0E9, Canada
| | - Sebastian Wachsmann-Hogiu
- Bioengineering, McGill University Faculty of Engineering, 3480 University St., MC362, Montreal, H3A 0E9l, Canada
| |
Collapse
|
37
|
Zhu F, Ji Y, Deng J, Li L, Bai X, Liu X, Lin B, Lu Y. Microfluidics-based technologies for the analysis of extracellular vesicles at the single-cell level and single-vesicle level. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.09.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
38
|
Horiguchi Y, Naono N, Sakamoto O, Takeuchi H, Yamaoka S, Miyahara Y. Methodology to Detect Biological Particles Using a Biosensing Surface Integrated in Resistive Pulse Sensing. ACS APPLIED MATERIALS & INTERFACES 2022; 14:20168-20178. [PMID: 35446533 DOI: 10.1021/acsami.1c25006] [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: 06/14/2023]
Abstract
Resistive pulse sensing (RPS) is an analytical method that can be used to individually count particles from a small sample. RPS simply monitors the physical characteristics of particles, such as size, shape, and charge density, and the integration of RPS with biosensing is an attractive theme to detect biological particles such as virus and bacteria. In this report, a methodology of biosensing on RPS was investigated. Polydopamine (PD), an adhesive component of mussels, was used as the base material to create a sensing surface. PD adheres to most materials, such as noble metals, metal oxides, semiconductors, and polymers; as a result, PD is a versatile intermediate layer for the fabrication of a biosensing surface. As an example of a biological particle, human influenza A virus (H1N1 subtype) was used to monitor translocation of particles through the pore membrane. When virus-specific ligands (6'-sialyllactose) were immobilized on the pore surface, the translocation time of the virus particles was considerably extended. The detailed translocation data suggest that the viral particles were trapped on the sensing surface by specific interactions. In addition, virus translocation processes on different pore surfaces were distinguished using machine learning. The result shows that the simple and versatile PD-based biosensor surface design was effective. This advanced RPS measurement system could be a promising analytical technique.
Collapse
Affiliation(s)
- Yukichi Horiguchi
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
| | - Norihiko Naono
- Aipore Inc., Cerulean Tower 15F, 26-1 Sakuragaokacho, Shibuya, Tokyo 150-8512, Japan
| | - Osamu Sakamoto
- Aipore Inc., Cerulean Tower 15F, 26-1 Sakuragaokacho, Shibuya, Tokyo 150-8512, Japan
| | - Hiroaki Takeuchi
- Department of Molecular Virology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo, Tokyo 113-8510, Japan
| | - Shoji Yamaoka
- Department of Molecular Virology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo, Tokyo 113-8510, Japan
| | - Yuji Miyahara
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
| |
Collapse
|
39
|
Dechantsreiter S, Ambrose AR, Worboys JD, Lim JME, Liu S, Shah R, Montero MA, Quinn AM, Hussell T, Tannahill GM, Davis DM. Heterogeneity in extracellular vesicle secretion by single human macrophages revealed by super-resolution microscopy. J Extracell Vesicles 2022; 11:e12215. [PMID: 35415881 PMCID: PMC9006015 DOI: 10.1002/jev2.12215] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/02/2022] [Accepted: 03/26/2022] [Indexed: 12/28/2022] Open
Abstract
The diverse origins, nanometre‐scale and invasive isolation procedures associated with extracellular vesicles (EVs) mean they are usually studied in bulk and disconnected from their parental cell. Here, we used super‐resolution microscopy to directly compare EVs secreted by individual human monocyte‐derived macrophages (MDMs). MDMs were differentiated to be M0‐, M1‐ or M2‐like, with all three secreting EVs at similar densities following activation. However, M0‐like cells secreted larger EVs than M1‐ and M2‐like macrophages. Proteomic analysis revealed variations in the contents of differently sized EVs as well as between EVs secreted by different MDM phenotypes. Super resolution microscopy of single‐cell secretions identified that the class II MHC protein, HLA‐DR, was expressed on ∼40% of EVs secreted from M1‐like MDMs, which was double the frequency observed for M0‐like and M2‐like EVs. Strikingly, human macrophages, isolated from the resected lungs of cancer patients, secreted EVs that expressed HLA‐DR at double the frequency and with greater intensity than M1‐like EVs. Quantitative analysis of single‐cell EV profiles from all four macrophage phenotypes revealed distinct secretion types, five of which were consistent across multiple sample cohorts. A sub‐population of M1‐like MDMs secreted EVs similar to lung macrophages, suggesting an expansion or recruitment of cells with a specific EV secretion profile within the lungs of cancer patients. Thus, quantitative analysis of EV heterogeneity can be used for single cell profiling and to reveal novel macrophage biology.
Collapse
Affiliation(s)
- Susanne Dechantsreiter
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ashley R Ambrose
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jonathan D Worboys
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Joey M E Lim
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Sylvia Liu
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Rajesh Shah
- Department of Cardiothoracic Surgery and Cellular Pathology, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - M Angeles Montero
- Cellular Pathology, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Anne Marie Quinn
- Department of Anatomic Pathology, University Hospital Galway, Galway, Ireland
| | - Tracy Hussell
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | | | - Daniel M Davis
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
40
|
Chen J, Tan Q, Yang Z, Jin Y. Engineered extracellular vesicles: potentials in cancer combination therapy. J Nanobiotechnology 2022; 20:132. [PMID: 35292030 PMCID: PMC8922858 DOI: 10.1186/s12951-022-01330-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/28/2022] [Indexed: 02/08/2023] Open
Abstract
Extracellular vesicles (EVs) are a group of secretory vesicles with cell-derived membrane and contents. Due to the cargo delivery capability, EVs can be designed as drug delivery platforms for cancer therapy. Biocompatibility and immune compatibility endow EVs with unique advantages compared with other nanocarriers. With the development of this field, multiple ingenious modification methods have been developed to obtain engineered EVs with desired performance. Application of engineered EVs in cancer therapy has gradually shifted from monotherapy to combinational therapy to fight against heterogeneous cancer cells and complex tumor microenvironment. In addition, the strong plasticity and load capacity of engineered EV make it potential to achieve various combinations of cancer treatment methods. In this review, we summarize the existing schemes of cancer combination therapy realized by engineered EVs, highlight the mechanisms and representative examples of these schemes and provide guidance for the future application of engineered EVs to design more effective cancer combination treatment plans.
Collapse
Affiliation(s)
- Jiangbin Chen
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, People's Republic of China
| | - Qi Tan
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, People's Republic of China
| | - Zimo Yang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, People's Republic of China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, People's Republic of China.
| |
Collapse
|
41
|
McNamara RP, Zhou Y, Eason AB, Landis JT, Chambers MG, Willcox S, Peterson TA, Schouest B, Maness NJ, MacLean AG, Costantini LM, Griffith JD, Dittmer DP. Imaging of surface microdomains on individual extracellular vesicles in 3-D. J Extracell Vesicles 2022; 11:e12191. [PMID: 35234354 PMCID: PMC8888793 DOI: 10.1002/jev2.12191] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/18/2022] [Accepted: 01/31/2022] [Indexed: 01/19/2023] Open
Abstract
Extracellular vesicles (EVs) are secreted from all cell types and are intimately involved in tissue homeostasis. They are being explored as vaccine and gene therapy platforms, as well as potential biomarkers. As their size is below the diffraction limit of light microscopy, direct visualizations have been daunting and single-particle studies under physiological conditions have been hampered. Here, direct stochastic optical reconstruction microscopy (dSTORM) was employed to visualize EVs in three-dimensions and to localize molecule clusters such as the tetraspanins CD81 and CD9 on the surface of individual EVs. These studies demonstrate the existence of membrane microdomains on EVs. These were confirmed by Cryo-EM. Individual particle visualization provided insights into the heterogeneity, structure, and complexity of EVs not previously appreciated.
Collapse
Affiliation(s)
- Ryan P. McNamara
- Department of Microbiology and ImmunologyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA,Lineberger Comprehensive Cancer CentreThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Yijun Zhou
- Department of Microbiology and ImmunologyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA,Lineberger Comprehensive Cancer CentreThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Anthony B. Eason
- Department of Microbiology and ImmunologyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA,Lineberger Comprehensive Cancer CentreThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Justin T. Landis
- Department of Microbiology and ImmunologyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA,Lineberger Comprehensive Cancer CentreThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Meredith G. Chambers
- Department of Microbiology and ImmunologyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA,Lineberger Comprehensive Cancer CentreThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Smaranda Willcox
- Lineberger Comprehensive Cancer CentreThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Tiffany A. Peterson
- Tulane National Primate Research CentreTulane UniversityCovingtonLouisianaUSA
| | - Blake Schouest
- Tulane National Primate Research CentreTulane UniversityCovingtonLouisianaUSA
| | - Nicholas J. Maness
- Tulane National Primate Research CentreTulane UniversityCovingtonLouisianaUSA
| | - Andrew G. MacLean
- Tulane National Primate Research CentreTulane UniversityCovingtonLouisianaUSA
| | - Lindsey M. Costantini
- Department of Biological and Biomedical SciencesNorth Carolina Central UniversityDurhamNorth CarolinaUSA
| | - Jack D. Griffith
- Lineberger Comprehensive Cancer CentreThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Dirk Peter Dittmer
- Department of Microbiology and ImmunologyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA,Lineberger Comprehensive Cancer CentreThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| |
Collapse
|
42
|
Soler-Botija C, Monguió-Tortajada M, Munizaga-Larroudé M, Gálvez-Montón C, Bayes-Genis A, Roura S. Mechanisms governing the therapeutic effect of mesenchymal stromal cell-derived extracellular vesicles: A scoping review of preclinical evidence. Biomed Pharmacother 2022; 147:112683. [DOI: 10.1016/j.biopha.2022.112683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/26/2022] [Accepted: 01/31/2022] [Indexed: 12/14/2022] Open
|
43
|
Engelbrecht L, Ollewagen T, de Swardt D. Advances in fluorescence microscopy can reveal important new aspects of tissue regeneration. Biochimie 2022; 196:194-202. [DOI: 10.1016/j.biochi.2022.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/19/2022] [Accepted: 02/02/2022] [Indexed: 12/12/2022]
|
44
|
Transmission Electron Microscopy as a Powerful Tool to Investigate the Interaction of Nanoparticles with Subcellular Structures. Int J Mol Sci 2021; 22:ijms222312789. [PMID: 34884592 PMCID: PMC8657944 DOI: 10.3390/ijms222312789] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/21/2021] [Accepted: 11/25/2021] [Indexed: 12/15/2022] Open
Abstract
Nanomedical research necessarily involves the study of the interactions between nanoparticulates and the biological environment. Transmission electron microscopy has proven to be a powerful tool in providing information about nanoparticle uptake, biodistribution and relationships with cell and tissue components, thanks to its high resolution. This article aims to overview the transmission electron microscopy techniques used to explore the impact of nanoconstructs on biological systems, highlighting the functional value of ultrastructural morphology, histochemistry and microanalysis as well as their fundamental contribution to the advancement of nanomedicine.
Collapse
|
45
|
Guo K, Li Z, Win A, Coreas R, Adkins GB, Cui X, Yan D, Cao M, Wang SE, Zhong W. Calibration-free analysis of surface proteins on single extracellular vesicles enabled by DNA nanostructure. Biosens Bioelectron 2021; 192:113502. [PMID: 34298496 PMCID: PMC8580803 DOI: 10.1016/j.bios.2021.113502] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/03/2021] [Accepted: 07/11/2021] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) are essential intercellular communicators that are of increasing interest as diagnostic biomarkers. Exploring their biological functions and clinical values, however, remains challenging due to their small sizes and high heterogeneity. Herein, we report an ultrasensitive method that employs target-initiated construction of DNA nanostructure to detect single EVs with an input as low as 100 vesicles/μL. Taking advantage of both DNA nanostructure labeling and EV membrane staining, the method can also permit calibration-free analysis of the protein profiles among different EV samples, leading to clear EV differentiation by their cell of origin. Moreover, this method allows co-localization of dual protein markers on the same EV, and the increased number of EVs carrying dual tumor proteins present in human serum could differentiate cancer patients at the early developmental stage from healthy controls. Our results demonstrate the great potential of this single-EV visualization method in non-invasive detection of the EV-based protein biomarkers for cancer diagnosis and treatment monitoring.
Collapse
Affiliation(s)
- Kaizhu Guo
- Department of Chemistry, University of California-Riverside, Riverside, CA, 92521, USA
| | - Zongbo Li
- Department of Chemistry, University of California-Riverside, Riverside, CA, 92521, USA
| | - Allison Win
- Department of Chemistry, University of California-Riverside, Riverside, CA, 92521, USA
| | - Roxana Coreas
- Environmental Toxicology Graduate Program, University of California-Riverside, Riverside, CA, 92521, USA
| | - Gary Brent Adkins
- Department of Chemistry, University of California-Riverside, Riverside, CA, 92521, USA
| | - Xinping Cui
- Department of Statistics, University of California-Riverside, Riverside, CA, 92521, USA
| | - Dong Yan
- Nanofabrication Facility, University of California-Riverside, Riverside, CA, 92521, USA
| | - Minghui Cao
- Department of Pathology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Shizhen Emily Wang
- Department of Pathology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Wenwan Zhong
- Department of Chemistry, University of California-Riverside, Riverside, CA, 92521, USA; Environmental Toxicology Graduate Program, University of California-Riverside, Riverside, CA, 92521, USA.
| |
Collapse
|
46
|
Min L, Wang B, Bao H, Li X, Zhao L, Meng J, Wang S. Advanced Nanotechnologies for Extracellular Vesicle-Based Liquid Biopsy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102789. [PMID: 34463056 PMCID: PMC8529441 DOI: 10.1002/advs.202102789] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Indexed: 05/09/2023]
Abstract
Extracellular vesicles (EVs) are emerging as a new source of biomarkers in liquid biopsy because of their wide presence in most body fluids and their ability to load cargoes from disease-related cells. Owing to the crucial role of EVs in disease diagnosis and treatment, significant efforts have been made to isolate, detect, and analyze EVs with high efficiency. A recent overview of advanced EV detection nanotechnologies is discussed here. First, several key challenges in EV-based liquid biopsies are introduced. Then, the related pivotal advances in nanotechnologies for EV isolation based on physical features, chemical affinity, and the combination of nanostructures and chemical affinity are summarized. Next, a summary of high-sensitivity sensors for EV detection and advanced approaches for single EV detection are provided. Later, EV analysis is introduced in practical clinical scenarios, and the application of machine learning in this field is highlighted. Finally, future opportunities for the development of next-generation nanotechnologies for EV detection are presented.
Collapse
Affiliation(s)
- Li Min
- Department of GastroenterologyBeijing Friendship HospitalCapital Medical UniversityNational Clinical Research Center for Digestive DiseasesBeijing Digestive Disease CenterBeijing Key Laboratory for Precancerous Lesion of Digestive DiseaseBeijing100050P. R. China
| | - Binshuai Wang
- Department of UrologyPeking University Third HospitalBeijing100191P. R. China
| | - Han Bao
- Key Laboratory of Bio‐inspired Materials and Interfacial ScienceCAS Center for Excellence in NanoscienceTechnical Institute of Physics and ChemistryChinese Academy of SciencesBeijing100190P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| | - Xinran Li
- Department of UrologyPeking University Third HospitalBeijing100191P. R. China
| | - Libo Zhao
- Echo Biotech Co., Ltd.Beijing102206P. R. China
| | - Jingxin Meng
- Key Laboratory of Bio‐inspired Materials and Interfacial ScienceCAS Center for Excellence in NanoscienceTechnical Institute of Physics and ChemistryChinese Academy of SciencesBeijing100190P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| | - Shutao Wang
- Key Laboratory of Bio‐inspired Materials and Interfacial ScienceCAS Center for Excellence in NanoscienceTechnical Institute of Physics and ChemistryChinese Academy of SciencesBeijing100190P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| |
Collapse
|
47
|
Fortunato D, Mladenović D, Criscuoli M, Loria F, Veiman KL, Zocco D, Koort K, Zarovni N. Opportunities and Pitfalls of Fluorescent Labeling Methodologies for Extracellular Vesicle Profiling on High-Resolution Single-Particle Platforms. Int J Mol Sci 2021; 22:10510. [PMID: 34638850 PMCID: PMC8508895 DOI: 10.3390/ijms221910510] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 12/26/2022] Open
Abstract
The relevance of extracellular vesicles (EVs) has grown exponentially, together with innovative basic research branches that feed medical and bioengineering applications. Such attraction has been fostered by the biological roles of EVs, as they carry biomolecules from any cell type to trigger systemic paracrine signaling or to dispose metabolism products. To fulfill their roles, EVs are transported through circulating biofluids, which can be exploited for the administration of therapeutic nanostructures or collected to intercept relevant EV-contained biomarkers. Despite their potential, EVs are ubiquitous and considerably heterogeneous. Therefore, it is fundamental to profile and identify subpopulations of interest. In this study, we optimized EV-labeling protocols on two different high-resolution single-particle platforms, the NanoFCM NanoAnalyzer (nFCM) and Particle Metrix ZetaView Fluorescence Nanoparticle Tracking Analyzer (F-NTA). In addition to the information obtained by particles' scattered light, purified and non-purified EVs from different cell sources were fluorescently stained with combinations of specific dyes and antibodies to facilitate their identification and characterization. Despite the validity and compatibility of EV-labeling strategies, they should be optimized for each platform. Since EVs can be easily confounded with similar-sized nanoparticles, it is imperative to control instrument settings and the specificity of staining protocols in order to conduct a rigorous and informative analysis.
Collapse
Affiliation(s)
| | - Danilo Mladenović
- HansaBioMed Life Sciences Ltd., 12618 Tallinn, Estonia; (D.M.); (F.L.); (K.-L.V.)
- School of Natural Sciences and Health, Tallinn University, 10120 Tallinn, Estonia;
| | | | - Francesca Loria
- HansaBioMed Life Sciences Ltd., 12618 Tallinn, Estonia; (D.M.); (F.L.); (K.-L.V.)
| | - Kadi-Liis Veiman
- HansaBioMed Life Sciences Ltd., 12618 Tallinn, Estonia; (D.M.); (F.L.); (K.-L.V.)
| | - Davide Zocco
- Exosomics SpA, 53100 Siena, Italy; (D.F.); (M.C.); (D.Z.)
- Cell and Gene Therapy Research and Development, Lonza Inc., Rockville, MD 20850, USA
| | - Kairi Koort
- School of Natural Sciences and Health, Tallinn University, 10120 Tallinn, Estonia;
| | - Natasa Zarovni
- Exosomics SpA, 53100 Siena, Italy; (D.F.); (M.C.); (D.Z.)
- HansaBioMed Life Sciences Ltd., 12618 Tallinn, Estonia; (D.M.); (F.L.); (K.-L.V.)
| |
Collapse
|
48
|
Nizamudeen ZA, Xerri R, Parmenter C, Suain K, Markus R, Chakrabarti L, Sottile V. Low-Power Sonication Can Alter Extracellular Vesicle Size and Properties. Cells 2021; 10:cells10092413. [PMID: 34572062 PMCID: PMC8466153 DOI: 10.3390/cells10092413] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/20/2021] [Accepted: 09/03/2021] [Indexed: 01/02/2023] Open
Abstract
Low-power sonication is widely used to disaggregate extracellular vesicles (EVs) after isolation, however, the effects of sonication on EV samples beyond dispersion are unclear. The present study analysed the characteristics of EVs collected from mesenchymal stem cells (MSCs) after sonication, using a combination of transmission electron microscopy, direct stochastic optical reconstruction microscopy, and flow cytometry techniques. Results showed that beyond the intended disaggregation effect, sonication using the lowest power setting available was enough to alter the size distribution, membrane integrity, and uptake of EVs in cultured cells. These results point to the need for a more systematic analysis of sonication procedures to improve reproducibility in EV-based cellular experiments.
Collapse
Affiliation(s)
| | - Rachael Xerri
- School of Pharmacy, The University of Nottingham, Nottingham NG7 2RD, UK; (R.X.); (C.P.); (K.S.)
| | - Christopher Parmenter
- School of Pharmacy, The University of Nottingham, Nottingham NG7 2RD, UK; (R.X.); (C.P.); (K.S.)
| | - Kiran Suain
- School of Pharmacy, The University of Nottingham, Nottingham NG7 2RD, UK; (R.X.); (C.P.); (K.S.)
| | - Robert Markus
- School of Life Sciences, The University of Nottingham, Nottingham NG7 2RD, UK;
| | - Lisa Chakrabarti
- School of Veterinary Medicine and Science, Sutton Bonington Campus, The University of Nottingham, Loughborough LE12 5RD, UK;
| | - Virginie Sottile
- School of Medicine, The University of Nottingham, Nottingham NG7 2RD, UK;
- Department of Molecular Medicine, The University of Pavia, 27100 Pavia, Italy
- Correspondence:
| |
Collapse
|
49
|
Lucas K, Dehghani M, Khire T, Gaborski T, Flax JD, Waugh RE, McGrath JL. A predictive model of nanoparticle capture on ultrathin nanoporous membranes. J Memb Sci 2021. [DOI: 10.1016/j.memsci.2021.119357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
50
|
Evaluation of Colon-Specific Plasma Nanovesicles as New Markers of Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13153905. [PMID: 34359806 PMCID: PMC8345452 DOI: 10.3390/cancers13153905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/22/2021] [Accepted: 07/29/2021] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Developing new and efficient approaches for the early diagnosis of colorectal cancer (CRC) is an important issue. Circulating extracellular nanovesicles (ENVs) present a promising class of cancer markers. Cells of well-differentiated adenocarcinomas retain the molecular characteristics of colon epithelial cells, and the ENVs secreted by these cells may have colon-specific surface markers. We hypothesize that an increase in the number of ENVs carrying colon-specific markers could serve as a diagnostic criterion for colorectal cancer. EXPERIMENTAL DESIGN Potential colon-specific markers were selected based on tissue-specific expression profile and cell surface membrane localization data. Plasma was collected from CRC patients (n = 48) and healthy donors (n = 50). The total population of ENVs was isolated with a two-phase polymer system. ENVs derived from colon epithelium cells were isolated using immune-beads with antibodies to colon-specific markers prior to labelling with antibodies against exosomal tetraspanins (CD63 and CD9) and quantification by flow cytometry. RESULTS The number of ENVs positive for single colon cancer markers was found to be significantly higher in the plasma of CRC patients compared with healthy donors. The efficacy of detection depends on the method of ENV labelling. The diagnostic efficacy was estimated by ROC analysis (the AUC varied between 0.71 and 0.79). The multiplexed isolation of colon-derived ENVs using immune-beads decorated with antibodies against five markers allowed for a further increase in the diagnostic potency of the method (AUC = 0.82). CONCLUSIONS ENVs derived from colon epithelium may serve as markers of differentiated CRC (adenocarcinomas). The composition of ligands used for capturing colon-derived ENVs and their method of labelling are critical for the efficacy of this proposed diagnostic approach.
Collapse
|