Extracellular Vesicles: Biology and Emerging Therapeutic Opportunities

A special issue of Pharmaceuticals (ISSN 1424-8247). This special issue belongs to the section "Pharmacology".

Deadline for manuscript submissions: closed (28 February 2022) | Viewed by 51004

Special Issue Editors


E-Mail Website
Guest Editor
Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
Interests: EV engineering; biotherpeutics delivery; RNA delivery

E-Mail Website
Co-Guest Editor
Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
Interests: EV therapy; Cell therapy; EV diagnostics; Immunology; Transfusion Medicine.

E-Mail Website
Co-Guest Editor
Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
Interests: Exosomes; Extracellular Vesicles; Flow Cytometry; Extracellular vesicle flow cytometry, Imaging Flow Cytometry, Extracellular Vesicle Engineering

E-Mail
Guest Editor
Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
Interests: liquid biopsies; RNA-sequencing; extracellular vesicles

Special Issue Information

Dear Colleagues,

Extracellular vesicles (EVs) are secreted by all cell types and can be found in all body fluids. EVs are intensely discussed and studied as potential new diagnostic biomarkers, and their ability to act as a natural vector offers a unique platform for the development of a new class of therapeutics. For example, unmodified EVs derived from mesenchymal stromal cells have demonstrated beneficial therapeutic effects in multiple animal disease models, and additionally EVs can be engineered to display various targeting and therapeutic moieties and shuttle cargo over biological barriers. Despite those prospects, many EV-related aspects related to EV production, isolation, formulation, cargo and function as well as their subsequent therapeutic application are yet poorly defined. This Special Issue has the aim to summarize the state-of-the-art, and the latest findings published within the field, as well as to elucidate future directions.

Dr. Samir EL Andaloussi
Dr. Joel Nordin
Dr. André Görgens
Dr. Daniel W. Hagey
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Pharmaceuticals is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Extracellular Vesicles
  • EVs, Exosomes
  • Microvesicles
  • EV engineering, protein engineering
  • EV cargo
  • vesicles
  • diagnostics
  • therapy

Related Special Issue

Published Papers (11 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

18 pages, 3448 KiB  
Article
Let-7i-5p Mediates the Therapeutic Effects of Exosomes from Human Placenta Choriodecidual Membrane-Derived Mesenchymal Stem Cells on Mitigating Endotoxin-Induced Mortality and Liver Injury in High-Fat Diet-Induced Obese Mice
by Chao-Yuan Chang, Kung-Yen Chen, Hung-Jen Shih, Milton Chiang, I-Tao Huang, Yen-Hua Huang and Chun-Jen Huang
Pharmaceuticals 2022, 15(1), 36; https://0-doi-org.brum.beds.ac.uk/10.3390/ph15010036 - 27 Dec 2021
Cited by 7 | Viewed by 3047
Abstract
Obesity complicates sepsis and increases the mortality of sepsis. We examined the effects of exosomes (from human placenta choriodecidual membrane-derived mesenchymal stem cells, pcMSCs) on preventing sepsis in obesity and the mitigating role of hsa-let-7i-5p microRNA. Obese mice (adult male C57BL/6J mice fed [...] Read more.
Obesity complicates sepsis and increases the mortality of sepsis. We examined the effects of exosomes (from human placenta choriodecidual membrane-derived mesenchymal stem cells, pcMSCs) on preventing sepsis in obesity and the mitigating role of hsa-let-7i-5p microRNA. Obese mice (adult male C57BL/6J mice fed a high-fat diet for 12 weeks) received normal saline (HFD), endotoxin (10 mg/kg, intraperitoneal (ip); HFDLPS), endotoxin with exosomes (1 × 108 particles/mouse, ip; HLE), or endotoxin with let-7i-5p microRNA inhibitor-pretreated exosomes (1 × 108 particles/mouse, ip; HLEi). Our data demonstrated that the 48-h survival rate in the HLE (100%) group was significantly higher than in the HFDLPS (50%) and HLEi (58.3%) groups (both p < 0.05). In the surviving mice, by contrast, levels of liver injury (injury score, plasma aspartate transaminase and alanine transaminase concentrations, tissue water content, and leukocyte infiltration in liver tissues; all p < 0.05), inflammation (nuclear factor-κB activation, hypoxia-inducible factor-1α activation, macrophage activation, and concentrations of tumor necrosis factor-α, interleukin-6, and leptin in liver tissues; all p < 0.05), and oxidation (malondialdehyde in liver tissues, with p < 0.001) in the HLE group were significantly lower than in the HFDLPS group. Levels of mitochondrial injury/dysfunction and apoptosis in liver tissues in the HLE group were also significantly lower than in the HFDLPS group (all p < 0.05). Inhibition of let-7i-5p microRNA offset the effects of the exosomes, with most of the aforementioned measurements in the HLEi group being significantly higher than in the HLE group (all p < 0.05). In conclusion, exosomes mitigated endotoxin-induced mortality and liver injury in obese mice, and these effects were mediated by let-7i-5p microRNA. Full article
Show Figures

Figure 1

25 pages, 4060 KiB  
Article
Antibodies Enhance the Suppressive Activity of Extracellular Vesicles in Mouse Delayed-Type Hypersensitivity
by Katarzyna Nazimek, Eugenio Bustos-Morán, Noelia Blas-Rus, Bernadeta Nowak, Justyna Totoń-Żurańska, Michał T. Seweryn, Paweł Wołkow, Olga Woźnicka, Rafał Szatanek, Maciej Siedlar, Philip W. Askenase, Francisco Sánchez-Madrid and Krzysztof Bryniarski
Pharmaceuticals 2021, 14(8), 734; https://0-doi-org.brum.beds.ac.uk/10.3390/ph14080734 - 27 Jul 2021
Cited by 6 | Viewed by 2632
Abstract
Previously, we showed that mouse delayed-type hypersensitivity (DTH) can be antigen-specifically downregulated by suppressor T cell-derived miRNA-150 carried by extracellular vesicles (EVs) that target antigen-presenting macrophages. However, the exact mechanism of the suppressive action of miRNA-150-targeted macrophages on effector T cells remained unclear, [...] Read more.
Previously, we showed that mouse delayed-type hypersensitivity (DTH) can be antigen-specifically downregulated by suppressor T cell-derived miRNA-150 carried by extracellular vesicles (EVs) that target antigen-presenting macrophages. However, the exact mechanism of the suppressive action of miRNA-150-targeted macrophages on effector T cells remained unclear, and our current studies aimed to investigate it. By employing the DTH mouse model, we showed that effector T cells were inhibited by macrophage-released EVs in a miRNA-150-dependent manner. This effect was enhanced by the pre-incubation of EVs with antigen-specific antibodies. Their specific binding to MHC class II-expressing EVs was proved in flow cytometry and ELISA-based experiments. Furthermore, by the use of nanoparticle tracking analysis and transmission electron microscopy, we found that the incubation of macrophage-released EVs with antigen-specific antibodies resulted in EVs’ aggregation, which significantly enhanced their suppressive activity in vivo. Nowadays, it is increasingly evident that EVs play an exceptional role in intercellular communication and selective cargo transfer, and thus are considered promising candidates for therapeutic usage. However, EVs appear to be less effective than their parental cells. In this context, our current studies provide evidence that antigen-specific antibodies can be easily used for increasing EVs’ biological activity, which has great therapeutic potential. Full article
Show Figures

Figure 1

13 pages, 3487 KiB  
Article
Engineered EVs for Oxidative Stress Protection
by Anna Maria Tolomeo, Santina Quarta, Alessandra Biasiolo, Mariagrazia Ruvoletto, Michela Pozzobon, Giada De Lazzari, Ricardo Malvicini, Cristian Turato, Giorgio Arrigoni, Patrizia Pontisso and Maurizio Muraca
Pharmaceuticals 2021, 14(8), 703; https://0-doi-org.brum.beds.ac.uk/10.3390/ph14080703 - 21 Jul 2021
Cited by 1 | Viewed by 2760
Abstract
Extracellular vesicles (EVs) are increasingly studied as vectors for drug delivery because they can transfer a variety of molecules across biological barriers. SerpinB3 is a serine protease inhibitor that has shown a protective anti-apoptotic function in a variety of stressful conditions. The aim [...] Read more.
Extracellular vesicles (EVs) are increasingly studied as vectors for drug delivery because they can transfer a variety of molecules across biological barriers. SerpinB3 is a serine protease inhibitor that has shown a protective anti-apoptotic function in a variety of stressful conditions. The aim of this study was to evaluate protection from oxidative stress-induced damage, using extracellular vesicles that overexpress SerpinB3 (EVs-SB3) in order to enhance the effect of extracellular vesicles on cellular homeostasis. EVs-SB3s were obtained from HepG2 cells engineered to overexpress SerpinB3 and they revealed significant proteomic changes, mostly characterized by a reduced expression of other proteins compared with EVs from non-engineered cells. These EV preparations showed a significantly higher protection from H2O2 induced oxidative stress in both the hepatoma cell line and in primary cardiomyocytes, compared to cells treated with naïve EVs or SerpinB3 alone, used at the same concentration. In conclusion, the induction of SerpinB3 transgene expression results in the secretion of EVs enriched with the protein product that exhibits enhanced cytoprotective activity, compared with naïve EVs or the nude SerpinB3 protein. Full article
Show Figures

Figure 1

23 pages, 5892 KiB  
Article
Equine Mesenchymal Stem/Stromal Cells Freeze-Dried Secretome (Lyosecretome) for the Treatment of Musculoskeletal Diseases: Production Process Validation and Batch Release Test for Clinical Use
by Michela Mocchi, Stefano Grolli, Silvia Dotti, Dario Di Silvestre, Riccardo Villa, Priscilla Berni, Virna Conti, Giulia Passignani, Francesca Brambilla, Maurizio Del Bue, Laura Catenacci, Milena Sorrenti, Lorena Segale, Elia Bari, Pierluigi Mauri, Maria Luisa Torre and Sara Perteghella
Pharmaceuticals 2021, 14(6), 553; https://0-doi-org.brum.beds.ac.uk/10.3390/ph14060553 - 10 Jun 2021
Cited by 10 | Viewed by 3006
Abstract
In the last decades, it has been demonstrated that the regenerative therapeutic efficacy of mesenchymal stromal cells is primarily due to the secretion of soluble factors and extracellular vesicles, collectively known as secretome. In this context, our work described the preparation and characterization [...] Read more.
In the last decades, it has been demonstrated that the regenerative therapeutic efficacy of mesenchymal stromal cells is primarily due to the secretion of soluble factors and extracellular vesicles, collectively known as secretome. In this context, our work described the preparation and characterization of a freeze-dried secretome (Lyosecretome) from adipose tissue-derived mesenchymal stromal cells for the therapy of equine musculoskeletal disorder. An intraarticular injectable pharmaceutical powder has been formulated, and the technological process has been validated in an authorized facility for veterinary clinical-use medicinal production. Critical parameters for quality control and batch release have been identified regarding (i) physicochemical properties; (ii) extracellular vesicle morphology, size distribution, and surface biomarker; (iii) protein and lipid content; (iv) requirements for injectable pharmaceutical dosage forms such as sterility, bacterial endotoxin, and Mycoplasma; and (v) in vitro potency tests, as anti-elastase activity and proliferative activity on musculoskeletal cell lines (tenocytes and chondrocytes) and mesenchymal stromal cells. Finally, proteins putatively responsible for the biological effects have been identified by Lyosecretome proteomic investigation: IL10RA, MXRA5, RARRES2, and ANXA1 modulate the inflammatory process RARRES2, NOD1, SERPINE1, and SERPINB9 with antibacterial activity. The work provides a proof-of-concept for the manufacturing of clinical-grade equine freeze-dried secretome, and prototypes are now available for safety and efficacy clinical trials in the treatment of equine musculoskeletal diseases Full article
Show Figures

Figure 1

16 pages, 2564 KiB  
Article
A Simple and Quick Method for Loading Proteins in Extracellular Vesicles
by Sara Busatto, Dalila Iannotta, Sierra A. Walker, Luisa Di Marzio and Joy Wolfram
Pharmaceuticals 2021, 14(4), 356; https://0-doi-org.brum.beds.ac.uk/10.3390/ph14040356 - 13 Apr 2021
Cited by 34 | Viewed by 5394
Abstract
Extracellular vesicles (EVs) mediate intercellular transport of biomolecular cargo in the body, making them promising delivery vehicles for bioactive compounds. Genetic engineering of producer cells has enabled encapsulation of therapeutic proteins in EVs. However, genetic engineering approaches can be expensive, time-consuming, and incompatible [...] Read more.
Extracellular vesicles (EVs) mediate intercellular transport of biomolecular cargo in the body, making them promising delivery vehicles for bioactive compounds. Genetic engineering of producer cells has enabled encapsulation of therapeutic proteins in EVs. However, genetic engineering approaches can be expensive, time-consuming, and incompatible with certain EV sources, such as human plasma and bovine milk. The goal of this study was to develop a quick, versatile, and simple method for loading proteins in EVs post-isolation. Proteins, including CRISPR associated protein 9 (Cas9), were bound to cationic lipids that were further complexed with MDA-MB-231 cell-derived EVs through passive incubation. Size-exclusion chromatography was used to remove components that were not complexed with EVs. The ability of EVs to mediate intracellular delivery of proteins was compared to conventional methods, such as electroporation and commercial protein transfection reagents. The results indicate that EVs retain native features following protein-loading and obtain similar levels of intracellular protein delivery as conventional methods, but display less toxicity. This method opens up opportunities for rapid exploration of EVs for protein delivery. Full article
Show Figures

Graphical abstract

10 pages, 2154 KiB  
Article
Assessment of Tumorigenic Potential in Mesenchymal-Stem/Stromal-Cell-Derived Small Extracellular Vesicles (MSC-sEV)
by Thong Teck Tan, Ruenn Chai Lai, Jayanthi Padmanabhan, Wei Kian Sim, Andre Boon Hwa Choo and Sai Kiang Lim
Pharmaceuticals 2021, 14(4), 345; https://0-doi-org.brum.beds.ac.uk/10.3390/ph14040345 - 09 Apr 2021
Cited by 26 | Viewed by 3084
Abstract
Mesenchymal-stem/stromal-cell-derived small extracellular vesicles (MSC-sEV) have been shown to ameliorate many diseases in preclinical studies. However, translating MSC-sEV into clinical use requires the development of scalable manufacturing processes for highly reproducible preparations of safe and potent MSC-sEVs. A major source of variability in [...] Read more.
Mesenchymal-stem/stromal-cell-derived small extracellular vesicles (MSC-sEV) have been shown to ameliorate many diseases in preclinical studies. However, translating MSC-sEV into clinical use requires the development of scalable manufacturing processes for highly reproducible preparations of safe and potent MSC-sEVs. A major source of variability in MSC-sEV preparations is EV producer cells. To circumvent variability in producer cells, clonal immortalized MSC lines as EV producer lines are increasingly being used for sEV production. The use of sEVs from immortalized producer cells inevitably raises safety concerns regarding the tumorigenicity or tumor promoting potential of the EV products. In this study, cells from E1-MYC line, a MSC cell line immortalized with the MYC gene, were injected subcutaneously into athymic nude mice. At 84 days post-injection, no tumor formation was observed at the injection site, lungs, or lymph nodes. E1-MYC cells pre-and post-sEV production did not exhibit anchorage-independent growth in soft agar. Daily intraperitoneal injections of 1 or 5 μg sEVs from E1-MYC into athymic nude mice with FaDu human head and neck cancer xenografts for 28 days did not promote or inhibit tumor growth relative to the xenograft treated with vehicle control. Therefore, MYC-immortalized MSCs are not tumorigenic and sEVs from these MSCs do not promote tumor growth. Full article
Show Figures

Figure 1

Review

Jump to: Research

20 pages, 2023 KiB  
Review
Extracellular Vesicles in Human Milk
by Yong Hu, Johannes Thaler and Rienk Nieuwland
Pharmaceuticals 2021, 14(10), 1050; https://0-doi-org.brum.beds.ac.uk/10.3390/ph14101050 - 15 Oct 2021
Cited by 25 | Viewed by 3888
Abstract
Milk supports the growth and development of infants. An increasing number of mostly recent studies have demonstrated that milk contains a hitherto undescribed component called extracellular vesicles (EVs). This presents questions regarding why milk contains EVs and what their function is. Recently, we [...] Read more.
Milk supports the growth and development of infants. An increasing number of mostly recent studies have demonstrated that milk contains a hitherto undescribed component called extracellular vesicles (EVs). This presents questions regarding why milk contains EVs and what their function is. Recently, we showed that EVs in human milk expose tissue factor, the protein that triggers coagulation or blood clotting, and that milk-derived EVs promote coagulation. Because bovine milk, which also contains EVs, completely lacks this coagulant activity, important differences are present in the biological functions of human milk-derived EVs between species. In this review, we will summarize the current knowledge regarding the presence and biochemical composition of milk EVs, their function(s) and potential clinical applications such as in probiotics, and the unique problems that milk EVs encounter in vivo, including survival of the gastrointestinal conditions encountered in the newborn. The main focus of this review will be human milk-derived EVs, but when available, we will also include information regarding non-human milk for comparison. Full article
Show Figures

Figure 1

39 pages, 4082 KiB  
Review
Extracellular Vesicles: A Double-Edged Sword in Sepsis
by Marlies Burgelman, Charysse Vandendriessche and Roosmarijn E. Vandenbroucke
Pharmaceuticals 2021, 14(8), 829; https://0-doi-org.brum.beds.ac.uk/10.3390/ph14080829 - 23 Aug 2021
Cited by 26 | Viewed by 4164
Abstract
Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to an infection. Several studies on mouse and patient sepsis samples have revealed that the level of extracellular vesicles (EVs) in the blood is altered compared to healthy controls, [...] Read more.
Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to an infection. Several studies on mouse and patient sepsis samples have revealed that the level of extracellular vesicles (EVs) in the blood is altered compared to healthy controls, but the different functions of EVs during sepsis pathology are not yet completely understood. Sepsis EVs are described as modulators of inflammation, lymphocyte apoptosis, coagulation and organ dysfunction. Furthermore, EVs can influence clinical outcome and it is suggested that EVs can predict survival. Both detrimental and beneficial roles for EVs have been described in sepsis, depending on the EV cellular source and the disease phase during which the EVs are studied. In this review, we summarize the current knowledge of EV sources and functions during sepsis pathology based on in vitro and mouse models, as well as patient samples. Full article
Show Figures

Graphical abstract

45 pages, 5569 KiB  
Review
Extracellular Vesicles in Skin Wound Healing
by Deimantė Narauskaitė, Gabrielė Vydmantaitė, Justina Rusteikaitė, Revathi Sampath, Akvilė Rudaitytė, Gabija Stašytė, María Isabel Aparicio Calvente and Aistė Jekabsone
Pharmaceuticals 2021, 14(8), 811; https://0-doi-org.brum.beds.ac.uk/10.3390/ph14080811 - 18 Aug 2021
Cited by 52 | Viewed by 10835
Abstract
Each year, millions of individuals suffer from a non-healing wound, abnormal scarring, or injuries accompanied by an infection. For these cases, scientists are searching for new therapeutic interventions, from which one of the most promising is the use of extracellular vesicles (EVs). Naturally, [...] Read more.
Each year, millions of individuals suffer from a non-healing wound, abnormal scarring, or injuries accompanied by an infection. For these cases, scientists are searching for new therapeutic interventions, from which one of the most promising is the use of extracellular vesicles (EVs). Naturally, EV-based signaling takes part in all four wound healing phases: hemostasis, inflammation, proliferation, and remodeling. Such an extensive involvement of EVs suggests exploiting their action to modulate the impaired healing phase. Furthermore, next to their natural wound healing capacity, EVs can be engineered for better defined pharmaceutical purposes, such as carrying specific cargo or targeting specific destinations by labelling them with certain surface proteins. This review aims to promote scientific awareness in basic and translational research of EVs by summarizing the current knowledge about their natural role in each stage of skin repair and the most recent findings in application areas, such as wound healing, skin regeneration, and treatment of dermal diseases, including the stem cell-derived, plant-derived, and engineered EVs. Full article
Show Figures

Graphical abstract

14 pages, 4620 KiB  
Review
Extracellular Vesicles and Their Potential Significance in the Pathogenesis and Treatment of Osteoarthritis
by Anne-Mari Mustonen and Petteri Nieminen
Pharmaceuticals 2021, 14(4), 315; https://0-doi-org.brum.beds.ac.uk/10.3390/ph14040315 - 01 Apr 2021
Cited by 26 | Viewed by 3473
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by inflammation, gradual destruction of articular cartilage, joint pain, and functional limitations that eventually lead to disability. Join tissues, including synovium and articular cartilage, release extracellular vesicles (EVs) that have been proposed to sustain joint [...] Read more.
Osteoarthritis (OA) is a chronic joint disease characterized by inflammation, gradual destruction of articular cartilage, joint pain, and functional limitations that eventually lead to disability. Join tissues, including synovium and articular cartilage, release extracellular vesicles (EVs) that have been proposed to sustain joint homeostasis as well as to contribute to OA pathogenesis. EVs transport biologically active molecules, and OA can be characterized by altered EV counts and composition in synovial fluid. Of EV cargo, specific non-coding RNAs could have future potential as diagnostic biomarkers for early OA. EVs may contribute to the propagation of inflammation and cartilage destruction by transporting and enhancing the production of inflammatory mediators and cartilage-degrading proteinases. In addition to inducing OA-related gene expression patterns in synoviocytes and articular chondrocytes, EVs can induce anti-OA effects, including increased extracellular matrix deposition and cartilage protection. Especially mesenchymal stem cell-derived EVs can alleviate intra-articular inflammation and relieve OA pain. In addition, surgically- or chemically-induced cartilage defects have been repaired with EV therapies in animal models. While human clinical trials are still in the future, the potential of actual cures to OA by EV products is very promising. Full article
Show Figures

Figure 1

27 pages, 1896 KiB  
Review
Matrix Vesicles: Role in Bone Mineralization and Potential Use as Therapeutics
by Sana Ansari, Bregje W. M. de Wildt, Michelle A. M. Vis, Carolina E. de Korte, Keita Ito, Sandra Hofmann and Yuana Yuana
Pharmaceuticals 2021, 14(4), 289; https://0-doi-org.brum.beds.ac.uk/10.3390/ph14040289 - 24 Mar 2021
Cited by 44 | Viewed by 6929
Abstract
Bone is a complex organ maintained by three main cell types: osteoblasts, osteoclasts, and osteocytes. During bone formation, osteoblasts deposit a mineralized organic matrix. Evidence shows that bone cells release extracellular vesicles (EVs): nano-sized bilayer vesicles, which are involved in intercellular communication by [...] Read more.
Bone is a complex organ maintained by three main cell types: osteoblasts, osteoclasts, and osteocytes. During bone formation, osteoblasts deposit a mineralized organic matrix. Evidence shows that bone cells release extracellular vesicles (EVs): nano-sized bilayer vesicles, which are involved in intercellular communication by delivering their cargoes through protein–ligand interactions or fusion to the plasma membrane of the recipient cell. Osteoblasts shed a subset of EVs known as matrix vesicles (MtVs), which contain phosphatases, calcium, and inorganic phosphate. These vesicles are believed to have a major role in matrix mineralization, and they feature bone-targeting and osteo-inductive properties. Understanding their contribution in bone formation and mineralization could help to target bone pathologies or bone regeneration using novel approaches such as stimulating MtV secretion in vivo, or the administration of in vitro or biomimetically produced MtVs. This review attempts to discuss the role of MtVs in biomineralization and their potential application for bone pathologies and bone regeneration. Full article
Show Figures

Figure 1

Back to TopTop