Zephyrnet Logo

Manufacture of extracellular vesicles derived from mesenchymal stromal cells

Date:

    • Caplan A.I.
    • Dennis J.E.

    Mesenchymal stem cells as trophic mediators.

    J. Cell. Biochem. 2006; 98: 1076-1084

    • Merckx G.
    • et al.

    Angiogenic effects of human dental pulp and bone marrow-derived mesenchymal stromal cells and their extracellular vesicles.

    Cells. 2020; 9: 312

    • Allen A.
    • et al.

    Mesenchymal stromal cell bioreactor for ex vivo reprogramming of human immune cells.

    Sci. Rep. 2020; 10: 10142

    • Hosseinkhani B.
    • et al.

    (Sub)populations of extracellular vesicles released by TNF-α-triggered human endothelial cells promote vascular inflammation and monocyte migration.

    J. Extracell. Vesicles. 2020; 91801153

    • Moon G.J.
    • et al.

    Application of mesenchymal stem cell-derived extracellular vesicles for stroke: Biodistribution and MicroRNA Study.

    Transl. Stroke Res. 2019; 10: 509-521

    • Alvarez-Erviti L.
    • et al.

    Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes.

    Nat. Biotechnol. 2011; 29: 341-345

    • Lee J.S.
    • et al.

    A long-term follow-up study of intravenous autologous mesenchymal stem cell transplantation in patients with ischemic stroke.

    Stem Cells. 2010; 28: 1099-1106

    • Kim S.J.
    • et al.

    Intravenous transplantation of mesenchymal stem cells preconditioned with early phase stroke serum: current evidence and study protocol for a randomized trial.

    Trials. 2013; 14: 317

    • Toma C.
    • et al.

    Fate of culture-expanded mesenchymal stem cells in the microvasculature: in vivo observations of cell kinetics.

    Circ. Res. 2009; 104: 398-402

    • Witwer K.W.
    • et al.

    Defining mesenchymal stromal cell (MSC)-derived small extracellular vesicles for therapeutic applications.

    J. Extracell. Vesicles. 2019; 81609206

    • Johnson J.
    • et al.

    From mesenchymal stromal cells to engineered extracellular vesicles: a new therapeutic paradigm.

    Front. Cell Dev. Biol. 2021; 9705676

    • Lenzini S.
    • et al.

    Matrix mechanics and water permeation regulate extracellular vesicle transport.

    Nat. Nanotechnol. 2020; 15: 217-223

    • Villar-Vesga J.
    • et al.

    Differential profile of systemic extracellular vesicles from sporadic and familial Alzheimer’s disease leads to neuroglial and endothelial cell degeneration.

    Front. Aging Neurosci. 2020; 12587989

    • Herrmann I.K.
    • et al.

    Extracellular vesicles as a next-generation drug delivery platform.

    Nat. Nanotechnol. 2021; 16: 748-759

    • Colao I.L.
    • et al.

    Manufacturing exosomes: a promising therapeutic platform.

    Trends Mol. Med. 2018; 24: 242-256

    • Paganini C.
    • et al.

    Scalable production and isolation of extracellular vesicles: available sources and lessons from current industrial bioprocesses.

    Biotechnol. J. 2019; 14e1800528

  • Exosome-based candidates move into the clinic.

    Nat. Rev. Drug Discov. 2021; 20: 6-7

    • Gimona M.
    • et al.

    Critical considerations for the development of potency tests for therapeutic applications of mesenchymal stromal cell-derived small extracellular vesicles.

    Cytotherapy. 2021; 23: 373-380

    • Gimona M.
    • et al.

    Manufacturing of human extracellular vesicle-based therapeutics for clinical use.

    Int. J. Mol. Sci. 2017; 18: 1190

    • Jossen V.
    • et al.

    Manufacturing human mesenchymal stem cells at clinical scale: process and regulatory challenges.

    Appl. Microbiol. Biotechnol. 2018; 102: 3981-3994

    • Haraszti R.A.
    • et al.

    Exosomes produced from 3D cultures of MSCs by tangential flow filtration show higher yield and improved activity.

    Mol. Ther. 2018; 26: 2838-2847

    • Watson D.C.
    • et al.

    Efficient production and enhanced tumor delivery of engineered extracellular vesicles.

    Biomaterials. 2016; 105: 195-205

    • Patel D.B.
    • et al.

    Enhanced extracellular vesicle production and ethanol-mediated vascularization bioactivity via a 3D-printed scaffold-perfusion bioreactor system.

    Acta Biomater. 2019; 95: 236-244

    • Saha B.
    • et al.

    MicroRNA cargo of extracellular vesicles from alcohol-exposed monocytes signals naive monocytes to differentiate into M2 macrophages.

    J. Biol. Chem. 2016; 291: 149-159

    • Guo S.
    • et al.

    Stimulating extracellular vesicles production from engineered tissues by mechanical forces.

    Nano Lett. 2021; 21: 2497-2504

    • Ambattu L.A.
    • et al.

    High frequency acoustic cell stimulation promotes exosome generation regulated by a calcium-dependent mechanism.

    Commun. Biol. 2020; 3: 553

    • Park K.-S.
    • et al.

    Enhancement of therapeutic potential of mesenchymal stem cell-derived extracellular vesicles.

    Stem Cell Res. Ther. 2019; 10: 288

    • Wolf M.
    • et al.

    A functional corona around extracellular vesicles enhances angiogenesis, skin regeneration and immunomodulation.

    J. Extracell. Vesicles. 2022; 11e12207

    • Li J.
    • et al.

    Serum-free culture alters the quantity and protein composition of neuroblastoma-derived extracellular vesicles.

    J. Extracell. Vesicles. 2015; 4: 26883

    • Gonzalez-King H.
    • et al.

    Hypoxia inducible factor-1α potentiates Jagged 1-mediated angiogenesis by mesenchymal stem cell-derived exosomes.

    Stem Cells. 2017; 35: 1747-1759

    • Zhang S.
    • et al.

    MSC exosomes mediate cartilage repair by enhancing proliferation, attenuating apoptosis and modulating immune reactivity.

    Biomaterials. 2018; 156: 16-27

    • Ma J.
    • et al.

    Exosomes derived from Akt-modified human umbilical cord mesenchymal stem cells improve cardiac regeneration and promote angiogenesis via activating platelet-derived growth factor D.

    Stem Cells Transl. Med. 2016; 6: 51-59

    • Mead B.
    • Tomarev S.

    Bone marrow-derived mesenchymal stem cells-derived exosomes promote survival of retinal ganglion cells through miRNA-dependent mechanisms.

    Stem Cells Transl. Med. 2017; 6: 1273-1285

    • Harting M.T.
    • et al.

    Inflammation-stimulated mesenchymal stromal cell-derived extracellular vesicles attenuate inflammation.

    Stem Cells. 2017; 36: 79-90

    • Zhu Y.
    • et al.

    Extracellular vesicles derived from human adipose-derived stem cells promote the exogenous angiogenesis of fat grafts via the let-7/AGO1/VEGF signalling pathway.

    Sci. Rep. 2020; 10: 5313

    • Wang J.
    • et al.

    Mesenchymal stem cell-derived extracellular vesicles alter disease outcomes via endorsement of macrophage polarization.

    Stem Cell Res. Ther. 2020; 11: 424

    • Whittaker T.E.
    • et al.

    Experimental artefacts can lead to misattribution of bioactivity from soluble mesenchymal stem cell paracrine factors to extracellular vesicles.

    J. Extracell. Vesicles. 2020; 91807674

    • Dominici M.
    • et al.

    Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement.

    Cytotherapy. 2006; 8: 315-317

    • Mucientes A.
    • et al.

    Influence of mesenchymal stem cell sources on their regenerative capacities on different surfaces.

    Cells. 2021; 10: 481

    • Kang I.
    • et al.

    Donor-dependent variation of human umbilical cord blood mesenchymal stem cells in response to hypoxic preconditioning and amelioration of limb ischemia.

    Exp. Mol. Med. 2018; 50: 1-15

    • Sathiyanathan P.
    • et al.

    A genomic biomarker that identifies human bone marrow-derived mesenchymal stem cells with high scalability.

    Stem Cells. 2020; 38: 1124-1136

    • Hu X.
    • et al.

    CRISPR/Cas9-mediated reversibly immortalized mouse bone marrow stromal stem cells (BMSCs) retain multipotent features of mesenchymal stem cells (MSCs).

    Oncotarget. 2017; 8: 111847-111865

    • Bloor A.J.C.
    • et al.

    Production, safety and efficacy of iPSC-derived mesenchymal stromal cells in acute steroid-resistant graft versus host disease: a phase I, multicenter, open-label, dose-escalation study.

    Nat. Med. 2020; 26: 1720-1725

    • Storm M.P.
    • et al.

    Hollow fiber bioreactors for in vivo-like mammalian tissue culture.

    J. Vis. Exp. 2016; 111: 53431

    • Bellio M.A.
    • et al.

    Systemic delivery of large-scale manufactured Wharton’s Jelly mesenchymal stem cell-derived extracellular vesicles improves cardiac function after myocardial infarction.

    J. Cardiovasc. Aging. 2022; 2: 9

    • Gobin J.
    • et al.

    Hollow-fiber bioreactor production of extracellular vesicles from human bone marrow mesenchymal stromal cells yields nanovesicles that mirrors the immuno-modulatory antigenic signature of the producer cell.

    Stem Cell Res. Ther. 2021; 12: 127

    • Patel D.B.
    • et al.

    Impact of cell culture parameters on production and vascularization bioactivity of mesenchymal stem cell-derived extracellular vesicles.

    Bioeng. Transl. Med. 2017; 2: 170-179

    • Yan L.
    • Wu X.

    Exosomes produced from 3D cultures of umbilical cord mesenchymal stem cells in a hollow-fiber bioreactor show improved osteochondral regeneration activity.

    Cell Biol. Toxicol. 2020; 36: 165-178

    • Mendt M.
    • et al.

    Generation and testing of clinical-grade exosomes for pancreatic cancer.

    JCI Insight. 2018; 3e99263

    • Grangier A.
    • et al.

    Technological advances towards extracellular vesicles mass production.

    Adv. Drug Deliv. Rev. 2021; 176113843

    • Silva A.K.A.
    • et al.

    Development of extracellular vesicle-based medicinal products: a position paper of the group “Extracellular Vesicle translatiOn to clinicaL perspectiVEs – EVOLVE France”.

    Adv. Drug Deliv. Rev. 2021; 179114001

    • Kumar A.
    • Starly B.

    Large scale industrialized cell expansion: producing the critical raw material for biofabrication processes.

    Biofabrication. 2015; 7044103

    • Si Y.
    • et al.

    Targeted exosomes for drug delivery: biomanufacturing, surface tagging, and validation.

    Biotechnol. J. 2020; 15e1900163

    • de Almeida Fuzeta M.
    • et al.

    Scalable production of human mesenchymal stromal cell-derived extracellular vesicles under serum-/xeno-free conditions in a microcarrier-based bioreactor culture system.

    Front. Cell Dev. Biol. 2020; 8553444

    • Dooley K.
    • et al.

    A versatile platform for generating engineered extracellular vesicles with defined therapeutic properties.

    Mol. Ther. 2021; 29: 1729-1743

    • Nie F.
    • et al.

    Extracellular vesicles derived from lipoaspirate fluid promote fat graft survival.

    Adipocyte. 2021; 10: 293-309

    • Tian M.
    • et al.

    Adipose-derived biogenic nanoparticles for suppression of inflammation.

    Small. 2020; 161904064

    • Busatto S.
    • et al.

    Tangential flow filtration for highly efficient concentration of extracellular vesicles from large volumes of fluid.

    Cells. 2018; 7: 273

    • Théry C.
    • et al.

    Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines.

    J. Extracell. Vesicles. 2018; 71535750

    • Lötvall J.
    • et al.

    Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles.

    J. Extracell. Vesicles. 2014; 3: 26913

    • Mwase C.
    • et al.

    Mechanical compression of human airway epithelial cells induces release of extracellular vesicles containing tenascin C.

    Cells. 2022; 11: 256

    • Chen T.
    • et al.

    Chemokine-containing exosomes are released from heat-stressed tumor cells via lipid raft-dependent pathway and act as efficient tumor vaccine.

    J. Immunol. 2011; 186: 2219-2228

    • Hedlund M.
    • et al.

    Thermal- and oxidative stress causes enhanced release of NKG2D ligand-bearing immunosuppressive exosomes in leukemia/lymphoma T and B cells.

    PLoS One. 2011; 6e16899

    • Zhao Z.
    • et al.

    Low-intensity ultrasound radiation increases exosome yield for efficient drug delivery.

    J. Drug Deliv. Sci. Technol. 2020; 57

    • Maredziak M.
    • et al.

    Static magnetic field enhances synthesis and secretion of membrane-derived microvesicles (MVs) rich in VEGF and BMP-2 in equine adipose-derived stromal cells (EqASCs)-a new approach in veterinary regenerative medicine.

    In Vitro Cell Dev. Biol. Anim. 2015; 51: 230-240

    • Fukuta T.
    • et al.

    Low level electricity increases the secretion of extracellular vesicles from cultured cells.

    Biochem. Biophys. Rep. 2020; 21100713

    • Ruan S.
    • et al.

    Light-induced high-efficient cellular production of immune functional extracellular vesicles.

    J. Extracell. Vesicles. 2022; 11e12194

    • Zhong J.-J.
    • et al.

    Enhancement of anthocyanin production by Perilla frutescens cells in a stirred bioreactor with internal light irradiation.

    J. Ferment. Bioeng. 1993; 75: 299-303

    • Wondraczek L.
    • et al.

    Biomimetic light dilution using side-emitting optical fiber for enhancing the productivity of microalgae reactors.

    Sci. Rep. 2019; 9: 9600

  • Gazeau, F. et al. Fluidic system for producing extracellular vesicles and associated method, WO/2019/002608

    • Parolini I.
    • et al.

    Microenvironmental pH is a key factor for exosome traffic in tumor cells.

    J. Biol. Chem. 2009; 284: 34211-34222

    • Han Y.
    • et al.

    Exosomes from hypoxia-treated human adipose-derived mesenchymal stem cells enhance angiogenesis through VEGF/VEGF-R.

    Int. J. Biochem. Cell Biol. 2019; 109: 59-68

    • Momen-Heravi F.
    • et al.

    Exosomes derived from alcohol-treated hepatocytes horizontally transfer liver specific miRNA-122 and sensitize monocytes to LPS.

    Sci. Rep. 2015; 5: 9991

    • Harmati M.
    • et al.

    Small extracellular vesicles convey the stress-induced adaptive responses of melanoma cells.

    Sci. Rep. 2019; 9: 15329

    • Park D.J.
    • et al.

    Improvement of stem cell-derived exosome release efficiency by surface-modified nanoparticles.

    J. Nanobiotechnol. 2020; 18: 178

    • Staubach S.
    • et al.

    Scaled preparation of extracellular vesicles from conditioned media.

    Adv. Drug Deliv. Rev. 2021; 177113940

    • Hagel L.
    • et al.

    Separation technologies.

    in: Hagel L. Handbook of Process Chromatography. 2nd edn. Academic Press, 2008: 81-125

    • Wang A.
    • et al.

    Comparison of different options for harvest of a therapeutic protein product from high cell density yeast fermentation broth.

    Biotechnol. Bioeng. 2006; 94: 91-104

    • Khadke S.
    • et al.

    Scalable solvent-free production of liposomes.

    J. Pharm. Pharmacol. 2020; 72: 1328-1340

    • Ungerechts G.
    • et al.

    Moving oncolytic viruses into the clinic: clinical-grade production, purification, and characterization of diverse oncolytic viruses.

    Mol. Ther. Methods Clin. Dev. 2016; 3: 16018

    • Gardiner C.
    • et al.

    Techniques used for the isolation and characterization of extracellular vesicles: results of a worldwide survey.

    J. Extracell. Vesicles. 2016; 5: 32945

    • Liangsupree T.
    • et al.

    Modern isolation and separation techniques for extracellular vesicles.

    J. Chromatogr. A. 2021; 1636461773

    • Weng Y.
    • et al.

    Effective isolation of exosomes with polyethylene glycol from cell culture supernatant for in-depth proteome profiling.

    Analyst. 2016; 141: 4640-4646

    • Zhang H.
    • et al.

    Improving isolation of extracellular vesicles by utilizing nanomaterials.

    Membranes (Basel). 2021; 12: 55

    • Paolini L.
    • et al.

    Residual matrix from different separation techniques impacts exosome biological activity.

    Sci. Rep. 2016; 6: 23550

    • Deregibus M.C.
    • et al.

    Charge-based precipitation of extracellular vesicles.

    Int. J. Mol. Med. 2016; 38: 1359-1366

    • Kowal J.
    • et al.

    Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes.

    Proc. Natl. Acad. Sci. U. S. A. 2016; 113: E968-E977

    • Veerman R.E.
    • et al.

    Molecular evaluation of five different isolation methods for extracellular vesicles reveals different clinical applicability and subcellular origin.

    J. Extracell. Vesicles. 2021; 10e12128

    • Kim D.K.
    • et al.

    Chromatographically isolated CD63+CD81+ extracellular vesicles from mesenchymal stromal cells rescue cognitive impairments after TBI.

    Proc. Natl. Acad. Sci. U. S. A. 2016; 113: 170-175

    • Balaj L.
    • et al.

    Heparin affinity purification of extracellular vesicles.

    Sci. Rep. 2015; 5: 10266

    • Cardoso R.M.S.
    • et al.

    Development of an optimized and scalable method for isolation of umbilical cord blood-derived small extracellular vesicles for future clinical use.

    Stem Cells Transl. Med. 2021; 10: 910-921

    • Zhang H.
    • et al.

    Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation.

    Nat. Cell Biol. 2018; 20: 332-343

    • Sitar S.
    • et al.

    Size characterization and quantification of exosomes by asymmetrical-flow field-flow fractionation.

    Anal. Chem. 2015; 87: 9225-9233

    • Multia E.
    • et al.

    Automated on-line isolation and fractionation system for nanosized biomacromolecules from human plasma.

    Anal. Chem. 2020; 92: 13058-13065

    • Roberts S.A.
    • et al.

    SPIN: rapid synthesis, purification, and concentration of small drug-loaded liposomes.

    J. Liposome Res. 2018; 28: 331-340

    • Liu F.
    • et al.

    The exosome total isolation chip.

    ACS Nano. 2017; 11: 10712-10723

    • Watson D.C.
    • et al.

    Scalable, cGMP-compatible purification of extracellular vesicles carrying bioactive human heterodimeric IL-15/lactadherin complexes.

    J. Extracell. Vesicles. 2018; 71442088

    • Kim K.
    • et al.

    Cyclic tangential flow filtration system for isolation of extracellular vesicles.

    APL Bioeng. 2021; 5016103

    • Ko S.Y.
    • Naora H.

    Extracellular vesicle membrane-associated proteins: emerging roles in tumor angiogenesis and anti-angiogenesis therapy resistance.

    Int. J. Mol. Sci. 2020; 21

    • Ko S.Y.
    • et al.

    Cancer-derived small extracellular vesicles promote angiogenesis by heparin-bound, bevacizumab-insensitive VEGF, independent of vesicle uptake.

    Commun. Biol. 2019; 2: 386

    • Yerneni S.S.
    • et al.

    Radioiodination of extravesicular surface constituents to study the biocorona, cell trafficking and storage stability of extracellular vesicles.

    Biochim. Biophys. Acta Gen. Subj. 2022; 1866130069

    • Chanda D.
    • et al.

    Fibronectin on the surface of extracellular vesicles mediates fibroblast invasion.

    Am. J. Respir. Cell Mol. Biol. 2019; 60: 279-288

    • Willis C.M.
    • et al.

    Extracellular vesicle fibrinogen induces encephalitogenic CD8+ T cells in a mouse model of multiple sclerosis.

    Proc. Natl. Acad. Sci. U. S. A. 2019; 116: 10488-10493

    • Park S.J.
    • et al.

    The effect of storage temperature on the biological activity of extracellular vesicles for the complement system.

    In Vitro Cell. Dev. Biol. Anim. 2018; 54: 423-429

    • Evtushenko E.G.
    • et al.

    Adsorption of extracellular vesicles onto the tube walls during storage in solution.

    PLoS One. 2020; 15e0243738

    • Tegegn T.Z.
    • et al.

    Characterization of procoagulant extracellular vesicles and platelet membrane disintegration in DMSO-cryopreserved platelets.

    J. Extracell. Vesicles. 2016; 5: 30422

    • Gelibter S.
    • et al.

    The impact of storage on extracellular vesicles: a systematic study.

    J. Extracell. Vesicles. 2022; 11e12162

    • Trenkenschuh E.
    • et al.

    Enhancing the stabilization potential of lyophilization for extracellular vesicles.

    Adv. Healthc. Mater. 2022; 112100538

    • Witwer K.W.
    • et al.

    Standardization of sample collection, isolation and analysis methods in extracellular vesicle research.

    J. Extracell. Vesicles. 2013; ()

    • Gorgens A.
    • et al.

    Identification of storage conditions stabilizing extracellular vesicles preparations.

    J. Extracell. Vesicles. 2022; 11e12238

    • Deville S.
    • et al.

    Comparison of extracellular vesicle isolation and storage methods using high-sensitivity flow cytometry.

    PLoS One. 2021; 16e0245835

    • Friedenstein A.J.
    • et al.

    Osteogenesis in transplants of bone marrow cells.

    Development. 1966; 16: 381-390

    • Harding C.
    • et al.

    Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes.

    J. Cell Biol. 1983; 97: 329-339

    • Pan B.-T.
    • Johnstone R.M.

    Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor.

    Cell. 1983; 33: 967-978

    • Kahn S.E.
    • et al.

    Purification of alkaline phosphatase from extracellular vesicles of fracture callus cartilage.

    Calcif. Tissue Res. 1978; 25: 85-92

    • Zitvogel L.
    • et al.

    Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell derived exosomes.

    Nat. Med. 1998; 4: 594-600

    • Pittenger M.F.
    • et al.

    Multilineage potential of adult human mesenchymal stem cells.

    Science. 1999; 284: 143-147

    • Timmers L.
    • et al.

    Reduction of myocardial infarct size by human mesenchymal stem cell conditioned medium.

    Stem Cell Res. 2008; 1: 129-137

    • Lai R.C.
    • et al.

    Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury.

    Stem Cell Res. 2010; 4: 214-222

    • Sun D.
    • et al.

    A novel nanoparticle drug delivery system: the anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes.

    Mol. Ther. 2010; 18: 1606-1614

    • Witwer K.W.
    • et al.

    Updating MISEV: evolving the minimal requirements for studies of extracellular vesicles.

    J. Extracell. Vesicles. 2021; 10e12182

    • Morse M.A.
    • et al.

    A phase I study of dexosome immunotherapy in patients with advanced non-small cell lung cancer.

    J. Transl. Med. 2005; 3: 9

    • Escudier B.
    • et al.

    Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: results of the first phase I clinical trial.

    J. Transl. Med. 2005; 3: 10

    • US Food and Drug Administration

    Guidance document. Potency tests for cellular and gene therapy products. Final guidance for industry: January 2011.

    • Nguyen V.V.T.
    • et al.

    Functional assays to assess the therapeutic potential of extracellular vesicles.

    J. Extracell. Vesicles. 2020; 10e12033

    • Reiner A.T.
    • et al.

    Concise review: developing best-practice models for the therapeutic use of extracellular vesicles.

    Stem Cells Transl. Med. 2017; 6: 1730-1739

    • Tieu A.
    • et al.

    An analysis of mesenchymal stem cell-derived extracellular vesicles for preclinical use.

    ACS Nano. 2020; 14: 9728-9743

    • Martinez Escudé
    • et al.

    Extracellular vesicles as a drug delivery system: a systematic review of preclinical studies.

    Adv. Drug Deliv. Rev. 2021; 175113801

    • Tahergorabi Z.
    • Khazaei M.

    A review on angiogenesis and its assays.

    Iran J. Basic Med. Sci. 2012; 15: 1110-1126

    • Huang Y.
    • et al.

    Bone marrow mesenchymal stem cell-derived exosomes promote rotator cuff tendon-bone healing by promoting angiogenesis and regulating M1 macrophages in rats.

    Stem Cell Res. Ther. 2020; 11: 496

    • Liang B.
    • et al.

    Dimethyloxaloylglycine-stimulated human bone marrow mesenchymal stem cell-derived exosomes enhance bone regeneration through angiogenesis by targeting the AKT/mTOR pathway.

    Stem Cell Res. Ther. 2019; 10: 335

    • Yang J.
    • et al.

    Umbilical cord-derived mesenchymal stem cell-derived exosomes combined pluronic F127 hydrogel promote chronic diabetic wound healing and complete skin regeneration.

    Int. J. Nanomedicine. 2020; 15: 5911-5926

    • Dabrowska S.
    • et al.

    Immunomodulatory and regenerative effects of mesenchymal stem cells and extracellular vesicles: therapeutic outlook for inflammatory and degenerative diseases.

    Front. Immunol. 2020; 11591065

    • Hirayama D.
    • et al.

    The phagocytic function of macrophage-enforcing innate immunity and tissue homeostasis.

    Int. J. Mol. Sci. 2017; 19: 92

    • Heo J.S.
    • et al.

    Adipose-derived mesenchymal stem cells promote M2 macrophage phenotype through exosomes.

    Stem Cells Int. 2019; 20197921760

    • Lo Sicco C.
    • et al.

    Mesenchymal stem cell-derived extracellular vesicles as mediators of anti-inflammatory effects: endorsement of macrophage polarization.

    Stem Cells Transl. Med. 2017; 6: 1018-1028

    • Paolicelli R.C.
    • et al.

    Microglia states and nomenclature: a field at its crossroads.

    Neuron. 2022; 110: 3458-3483

    • Suh J.H.
    • et al.

    Therapeutic application of exosomes in inflammatory diseases.

    Int. J. Mol. Sci. 2021; 22: 1144

    • Guy R.
    • Offen D.

    Promising opportunities for treating neurodegenerative diseases with mesenchymal stem cell-derived exosomes.

    Biomolecules. 2020; 10: 1320

    • Lopez-Verrilli M.A.
    • et al.

    Mesenchymal stem cell-derived exosomes from different sources selectively promote neuritic outgrowth.

    Neuroscience. 2016; 320: 129-139

    • Ching R.C.
    • et al.

    Schwann cell-like differentiated adipose stem cells promote neurite outgrowth via secreted exosomes and RNA transfer.

    Stem Cell Res. Ther. 2018; 9: 266

    • Geeurickx E.
    • et al.

    The generation and use of recombinant extracellular vesicles as biological reference material.

    Nat. Commun. 2019; 10: 3288

    • Wörle-Knirsch J.M.
    • et al.

    Oops they did it again! Carbon nanotubes hoax scientists in viability assays.

    Nano Lett. 2006; 6: 1261-1268

    • Álvarez V.
    • et al.

    The immunomodulatory activity of extracellular vesicles derived from endometrial mesenchymal stem cells on CD4+ T cells is partially mediated by TGFbeta.

    J. Tissue Eng. Regen. Med. 2018; 12: 2088-2098

    • Oszvald A.
    • et al.

    Extracellular vesicles transmit epithelial growth factor activity in the intestinal stem cell niche.

    Stem Cells. 2020; 38: 291-300

    • Ke X.
    • et al.

    Esophageal Adenocarcinoma-derived extracellular vesicle microRNAs induce a neoplastic phenotype in gastric organoids.

    Neoplasia. 2017; 19: 941-949

    • Gómez-Cid L.
    • et al.

    The essential need for a validated potency assay for cell-based therapies in cardiac regenerative and reparative medicine. A practical approach to test development.

    Stem Cell Rev. Rep. 2021; 17: 2235-2244

    • Kim H.
    • et al.

    Identification of molecules responsible for therapeutic effects of extracellular vesicles produced from iPSC-derived MSCs on Sjögren’s syndrome.

    Aging Dis. 2021; 12: 1409-1422

    • Williams A.M.
    • et al.

    Mesenchymal stem cell-derived exosomes provide neuroprotection and improve long-term neurologic outcomes in a swine model of traumatic brain injury and hemorrhagic shock.

    J. Neurotrauma. 2019; 36: 54-60

    • Potter D.R.
    • et al.

    Mesenchymal stem cell-derived extracellular vesicles attenuate pulmonary vascular permeability and lung injury induced by hemorrhagic shock and trauma.

    J. Trauma Acute Care Surg. 2018; 84: 245-256

    • Garcia N.A.
    • et al.

    Glucose starvation in cardiomyocytes enhances exosome secretion and promotes angiogenesis in endothelial cells.

    PLoS One. 2015; 10e0138849

    • Kim W.S.
    • et al.

    Comparison of exosomes derived from non- and gamma-irradiated melanoma cancer cells as a potential antigenic and immunogenic source for dendritic cell-based immunotherapeutic vaccine.

    Vaccines (Basel). 2020; 8: 699

  • spot_img

    Latest Intelligence

    spot_img