Isolation methods of exosomes derived from dental stem cells

Paras Ahmad , Nathan Estrin , Nima Farshidfar , Yufeng Zhang , Richard J. Miron

International Journal of Oral Science ›› 2025, Vol. 17 ›› Issue (1) : 50

PDF
International Journal of Oral Science ›› 2025, Vol. 17 ›› Issue (1) : 50 DOI: 10.1038/s41368-025-00370-y
Review Article

Isolation methods of exosomes derived from dental stem cells

Author information +
History +
PDF

Abstract

Mesenchymal stem cells are highly regarded for their potential in tissue repair and regenerative medicine due to their multipotency and self-renewal abilities. Recently, mesenchymal stem cells have been redefined as “medical signaling cells,” with their primary biological effects mediated through exosome secretion. These exosomes, which contain lipids, proteins, RNA, and metabolites, are crucial in regulating various biological processes and enhancing regenerative therapies. Exosomes replicate the effects of their parent cells while offering benefits such as reduced side effects, low immunogenicity, excellent biocompatibility, and high drug-loading capacity. Dental stem cells, including those from apical papilla, gingiva, dental pulp, and other sources, are key contributors to exosome-mediated regenerative effects, such as tumor cell apoptosis, neuroprotection, angiogenesis, osteogenesis, and immune modulation. Despite their promise, clinical application of exosomes is limited by challenges in isolation techniques. Current methods face issues of complexity, inefficiency, and insufficient purity, hindering detailed analysis. Recent advancements, such as micro-electromechanical systems, alternating current electroosmosis, and serum-free three-dimensional cell cultures, have improved exosome isolation efficacy. This review synthesizes nearly 200 studies on dental stem cell-derived exosomes, highlighting their potential in treating a wide range of conditions, including periodontal diseases, cancer, neurodegenerative disorders, diabetes, and more. Optimized isolation methods offer a path forward for overcoming current limitations and advancing the clinical use of exosome-based therapies.

Cite this article

Download citation ▾
Paras Ahmad, Nathan Estrin, Nima Farshidfar, Yufeng Zhang, Richard J. Miron. Isolation methods of exosomes derived from dental stem cells. International Journal of Oral Science, 2025, 17(1): 50 DOI:10.1038/s41368-025-00370-y

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

RaposoG, StoorvogelW. Extracellular vesicles: exosomes, microvesicles, and friends. J. Cell Biol., 2013, 200: 373-383

[2]

BeerKB, WehmanAM. Mechanisms and functions of extracellular vesicle release in vivo—what we can learn from flies and worms. Cell Adhes. Migr., 2017, 11: 135-150

[3]

MironRJ, ZhangY. Understanding exosomes: Part 1—Characterization, quantification and isolation techniques. Periodontology 2000, 2024, 94: 231-256

[4]

MironRJ, EstrinNE, SculeanA, ZhangY. Understanding exosomes: Part 2—Emerging leaders in regenerative medicine. Periodontology 2000, 2024, 94: 257-414

[5]

MironRJ, EstrinNE, SculeanA, ZhangY. Understanding exosomes: Part 3—therapeutic+ diagnostic potential in dentistry. Periodontology 2000, 2024, 94: 415-482

[6]

HadeMD, SuireCN, SuoZ. Mesenchymal stem cell-derived exosomes: applications in regenerative medicine. Cells, 2021, 10: 1959

[7]

HuW, WangW, ChenZ, ChenY, WangZ. Engineered exosomes and composite biomaterials for tissue regeneration. Theranostics, 2024, 14: 2099

[8]

TanF, et al.. Clinical applications of stem cell-derived exosomes. Signal Transduct. Target. Ther., 2024, 9: 17

[9]

BojicS, VolarevicV, LjujicB, StojkovicM. Dental stem cells-characteristics and potential. World Journal of Stem Cells, 2014, 13: 1610-1624

[10]

NakajimaK, et al.. Comparison of the bone regeneration ability between stem cells from human exfoliated deciduous teeth, human dental pulp stem cells and human bone marrow mesenchymal stem cells. Biochem. Biophys. Res. Commun., 2018, 497: 876-882

[11]

SonoyamaW, et al.. Mesenchymal stem cell-mediated functional tooth regeneration in swine. PLoS ONE, 2006, 1: e79

[12]

MiuraM, et al.. SHED: stem cells from human exfoliated deciduous teeth. Proc. Natl Acad. Sci., 2003, 100: 5807-5812

[13]

HandaK, et al.. Progenitor cells from dental follicle are able to form cementum matrix in vivo. Connect. Tissue Res., 2002, 43: 406-408

[14]

ZhangQ, et al.. Mesenchymal stem cells derived from human gingiva are capable of immunomodulatory functions and ameliorate inflammation-related tissue destruction in experimental colitis. J. Immunol., 2009, 183: 7787-7798

[15]

SeoB-M, et al.. Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet, 2004, 364: 149-155

[16]

GronthosS, MankaniM, BrahimJ, RobeyPG, ShiS. Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc. Natl Acad. Sci., 2000, 97: 13625-13630

[17]

AlyLAA. Stem cells: Sources, and regenerative therapies in dental research and practice. World J. Stem Cells, 2015, 7: 1047

[18]

ChalisserryEP, NamSY, ParkSH, AnilS. Therapeutic potential of dental stem cells. J. Tissue Eng., 2017, 8: 2041731417702531

[19]

NutiN, CoralloC, ChanB, FerrariM, Gerami-NainiB. Multipotent differentiation of human dental pulp stem cells: a literature review. Stem Cell Rev. Rep., 2016, 12: 511-523

[20]

SharpePT. Dental mesenchymal stem cells. Development, 2016, 143: 2273-2280

[21]

CampanellaV. Dental stem cells: current research and future applications. Eur. J. Paediatr. Dent., 2018, 19: 257-

[22]

SuiB, et al.. Dental pulp stem cells: from discovery to clinical application. J. Endod., 2020, 46: S46-S55

[23]

YamadaY, Nakamura-YamadaS, KusanoK, BabaS. Clinical potential and current progress of dental pulp stem cells for various systemic diseases in regenerative medicine: a concise review. Int. J. Mol. Sci., 2019, 20: 1132

[24]

LiY, DuanX, ChenY, LiuB, ChenG. Dental stem cell-derived extracellular vesicles as promising therapeutic agents in the treatment of diseases. Int. J. Oral. Sci., 2022, 14: 2

[25]

SyromiatnikovaV, ProkopevaA, GomzikovaM. Methods of the large-scale production of extracellular vesicles. Int. J. Mol. Sci., 2022, 23: 10522

[26]

ChenH, et al.. Exosomes, a new star for targeted delivery. Front. Cell Dev. Biol., 2021, 9: 751079

[27]

HeJ, et al.. Exosomal targeting and its potential clinical application. Drug Deliv. Transl. Res., 2022, 12: 2385-2402

[28]

MaiZ, et al.. Translational and clinical applications of dental stem cell-derived exosomes. Front. Genet., 2021, 12: 750990

[29]

SchepiciG, SilvestroS, MazzonE. Regenerative effects of exosomes-derived MSCs: an overview on spinal cord injury experimental studies. Biomedicines, 2023, 11: 201

[30]

NingX, et al.. Dental stem cell-derived exosomes: a review of their isolation, classification, functions, and mechanisms. Stem Cells Int., 2024, 2024: 2187392

[31]

RenS, LinY, LiuW, YangL, ZhaoM. MSC-Exos: important active factor of bone regeneration. Front. Bioeng. Biotechnol., 2023, 11: 1136453

[32]

AbdullahMF, et al.. Proliferation rate of stem cells derived from human dental pulp and identification of differentially expressed genes. Cell Biol. Int., 2014, 38: 582-590

[33]

Al-MaswaryAA, et al.. Exploring the neurogenic differentiation of human dental pulp stem cells. PLoS ONE, 2022, 17: e0277134

[34]

ZhouL, ZhaoS, XingX. Effects of different signaling pathways on odontogenic differentiation of dental pulp stem cells: a review. Front. Physiol., 2023, 14: 1272764

[35]

ZouJ, et al.. Exosomes derived from odontogenic stem cells: its role in the dentin-pulp complex. Regenerative Ther., 2023, 24: 135-146

[36]

BrunelloG, et al.. Exosomes derived from dental pulp stem cells show different angiogenic and osteogenic properties in relation to the age of the donor. Pharmaceutics, 2022, 14: 908

[37]

ShenZ, et al.. Effects of mesenchymal stem cell-derived exosomes on autoimmune diseases. Front. Immunol., 2021, 12: 749192

[38]

LiP, OuQ, ShiS, ShaoC. Immunomodulatory properties of mesenchymal stem cells/dental stem cells and their therapeutic applications. Cell. Mol. Immunol., 2023, 20: 558-569

[39]

KimHI, et al.. Recent advances in extracellular vesicles for therapeutic cargo delivery. Exp. Mol. Med., 2024, 56: 836-849

[40]

LiangY, et al.. Cell-derived nanovesicle-mediated drug delivery to the brain: principles and strategies for vesicle engineering. Mol. Ther., 2023, 31: 1207-1224

[41]

KibriaG, RamosEK, WanY, GiusDR, LiuH. Exosomes as a drug delivery system in cancer therapy: potential and challenges. Mol. Pharmaceutics, 2018, 15: 3625-3633

[42]

LuanX, et al.. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacologica Sin., 2017, 38: 754-763

[43]

HanY, et al.. Overview and update on methods for cargo loading into extracellular vesicles. Processes, 2021, 9: 356

[44]

KohHB, KimHJ, KangS-W, YooT-H. Exosome-based drug delivery: translation from bench to clinic. Pharmaceutics, 2023, 15: 2042

[45]

DuanX, et al.. A new subtype of artificial cell-derived vesicles from dental pulp stem cells with the bioequivalence and higher acquisition efficiency compared to extracellular vesicles. J. Extracell. Vesicles, 2024, 13: e12473

[46]

KurianTK, BanikS, GopalD, ChakrabartiS, MazumderN. Elucidating methods for isolation and quantification of exosomes: a review. Mol. Biotechnol., 2021, 63: 249-266

[47]

AlzhraniGN, et al.. Exosomes: Isolation, characterization, and biomedical applications. Cell Biol. Int., 2021, 45: 1807-1831

[48]

LiP, KaslanM, LeeSH, YaoJ, GaoZ. Progress in exosome isolation techniques. Theranostics, 2017, 7: 789

[49]

LinS, et al.. Progress in microfluidics-based exosome separation and detection technologies for diagnostic applications. Small, 2020, 16: 1903916

[50]

KamerkarS, et al.. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature, 2017, 546: 498-503

[51]

DingL, et al.. A holistic review of the state-of-the-art microfluidics for exosome separation: an overview of the current status, existing obstacles, and future outlook. Small, 2021, 17: 2007174

[52]

MohammadiM, et al.. Emerging technologies and commercial products in exosome-based cancer diagnosis and prognosis. Biosens. Bioelectron., 2021, 183: 113176

[53]

CoumansFA, et al.. Methodological guidelines to study extracellular vesicles. Circulation Res., 2017, 120: 1632-1648

[54]

ChenJ, et al.. Review on strategies and technologies for exosome isolation and purification. Front. Bioeng. Biotechnol., 2022, 9: 811971

[55]

WuM, et al.. Separating extracellular vesicles and lipoproteins via acoustofluidics. Lab Chip, 2019, 19: 1174-1182

[56]

SódarBW, et al.. Low-density lipoprotein mimics blood plasma-derived exosomes and microvesicles during isolation and detection. Sci. Rep., 2016, 6 ArticleID: 24316

[57]

KarimiN, et al.. Detailed analysis of the plasma extracellular vesicle proteome after separation from lipoproteins. Cell. Mol. Life Sci., 2018, 75: 2873-2886

[58]

LudwigN, WhitesideTL, ReichertTE. Challenges in exosome isolation and analysis in health and disease. Int. J. Mol. Sci., 2019, 20: 4684

[59]

HuaS, et al.. Periodontal and dental pulp cell-derived small extracellular vesicles: a review of the current status. Nanomaterials, 2021, 11: 1858

[60]

LiuC, SuC. Design strategies and application progress of therapeutic exosomes. Theranostics, 2019, 9: 1015

[61]

ThéryC, 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, 7: 1535750

[62]

KonoshenkoMY, LekchnovEA, VlassovAV, LaktionovPP. Isolation of extracellular vesicles: general methodologies and latest trends. BioMed. Res. Int., 2018, 2018: 8545347

[63]

SafdarA, SaleemA, TarnopolskyMA. The potential of endurance exercise-derived exosomes to treat metabolic diseases. Nat. Rev. Endocrinol., 2016, 12: 504-517

[64]

ZhangT, et al.. Extracellular vesicles derived from human dental mesenchymal stem cells stimulated with low-intensity pulsed ultrasound alleviate inflammation-induced bone loss in a mouse model of periodontitis. Genes Dis., 2023, 10: 1613-1625

[65]

LiuD, ShiB, ZhouW, TaoG. Exosomes from hypoxia-conditioned apical papilla stem cells accelerate angiogenesis in vitro through Notch/JAG1/VEGF signaling. Tissue Cell, 2023, 84: 102197

[66]

LinX, WangH, WuT, ZhuY, JiangL. Exosomes derived from stem cells from apical papilla promote angiogenesis via miR-126 under hypoxia. Oral. Dis., 2023, 29: 3408-3419

[67]

NieY-F, et al.. Apical papilla stem cell-derived exosomes regulate lipid metabolism and alleviate inflammation in the MCD-induced mouse NASH model. Biochem. Pharmacol., 2024, 222: 116073

[68]

JingX, et al.. Dynamically bioresponsive DNA hydrogel incorporated with dual-functional stem cells from apical papilla-derived exosomes promotes diabetic bone regeneration. ACS Appl. Mater. Interfaces, 2022, 14: 16082-16099

[69]

YuS, et al.. Exosomes derived from stem cells from the apical papilla alleviate inflammation in rat pulpitis by upregulating regulatory T cells. Int. Endod. J., 2022, 55: 517-530

[70]

WangA, et al.. Identification and comparison of piRNA expression profiles of exosomes derived from human stem cells from the apical papilla and bone marrow mesenchymal stem cells. Stem Cells Dev., 2020, 29: 511-520

[71]

HadadyH, KaramaliF, EjeianF, SoroushzadehS, Nasr-EsfahaniMH. Potential neuroprotective effect of stem cells from apical papilla derived extracellular vesicles enriched by lab-on-chip approach during retinal degeneration. Cell. Mol. Life Sci., 2022, 79: 350

[72]

HadadyH, et al.. AC electrokinetic isolation and detection of extracellular vesicles from dental pulp stem cells: Theoretical simulation incorporating fluid mechanics. Electrophoresis, 2021, 42: 2018-2026

[73]

HuangT-Y, ChienM-S, SuW-T. Therapeutic potential of pretreatment with exosomes derived from stem cells from the apical papilla against cisplatin-induced acute kidney injury. Int. J. Mol. Sci., 2022, 23: 5721

[74]

LiuY, et al.. Exosomes derived from stem cells from apical papilla promote craniofacial soft tissue regeneration through enhancing Cdc42-mediated vascularization. Stem Cell Research and Therapy, 2020, 12: 76

[75]

ZhuangX, et al.. Exosomes derived from stem cells from the apical papilla promote dentine-pulp complex regeneration by inducing specific dentinogenesis. Stem Cells Int., 2020, 2020: 5816723

[76]

WangH, et al.. Odontoblastic exosomes attenuate apoptosis in neighboring cells. J. Dent. Res., 2019, 98: 1271-1278

[77]

YangS, et al.. Extracellular vesicles delivering nuclear factor I/C for hard tissue engineering: treatment of apical periodontitis and dentin regeneration. J. Tissue Eng., 2022, 13: 20417314221084095

[78]

Gratpain, V. et al. Influence of a pro-inflammatory stimulus on the miRNA and lipid content of human dental stem cell-derived extracellular vesicles and their impact on microglial activation. Heliyon 10 (2024).

[79]

LiM, et al.. A ROS-responsive hydrogel incorporated with dental follicle stem cell-derived small extracellular vesicles promotes dental pulp repair by ameliorating oxidative stress. Bioact. Mater., 2024, 36: 524-540

[80]

FuH, et al.. Mesenchymal stem cells-derived extracellular vesicles protect against oxidative stress-induced xenogeneic biological root injury via adaptive regulation of the PI3K/Akt/NRF2 pathway. J. Nanobiotechnol., 2023, 21 ArticleID: 466

[81]

Liang, L. et al. High-yield nanovesicles extruded from dental follicle stem cells promote the regeneration of periodontal tissues as an alternative of exosomes. J. Clin. Periodontol. 51, 1395–1407 (2024).

[82]

Huang, Y. et al. Lipopolysaccharide-preconditioned dental follicle stem cells derived small extracellular vesicles treating periodontitis via reactive oxygen species/mitogen-activated protein kinase signaling-mediated antioxidant effect. Int. J. Nanomed. 17, 799–819 (2022).

[83]

MaoE, et al.. Human dental follicle cell-derived small extracellular vesicles attenuate temporomandibular joint cartilage damage through inhibiting HIF-2α. J. Tissue Eng. Regenerative Med., 2023, 2023: 6625123

[84]

MaL, et al.. Small extracellular vesicles from dental follicle stem cells provide biochemical cues for periodontal tissue regeneration. Stem Cell Res. Ther., 2022, 13: 92

[85]

YiG, et al.. Matrix vesicles from dental follicle cells improve alveolar bone regeneration via activation of the PLC/PKC/MAPK pathway. Stem Cell Res. Ther., 2022, 13: 41

[86]

DengY, LiuZ, LuM. Extracellular vesicles deviced from hypoxia-3D-GMSCs rescue the mitochondrial dysfunction of aging-GMSCs. Biochem. Biophys. Res. Commun., 2024, 717: 150021

[87]

YuZ, et al.. TNF-α stimulation enhances the neuroprotective effects of gingival MSCs derived exosomes in retinal ischemia-reperfusion injury via the MEG3/miR-21a-5p axis. Biomaterials, 2022, 284: 121484

[88]

ZarubovaJ, et al.. Engineered delivery of dental stem-cell-derived extracellular vesicles for periodontal tissue regeneration. Adv. Healthc. Mater., 2022, 11: 2102593

[89]

LiangX, et al.. Nanoscale exosomes derived from gingiva mesenchymal stem cells for radiotherapy-induced apoptosis in non-small cell lung cancer cells. ACS Appl. Nano Mater., 2023, 6: 13533-13542

[90]

HuY, et al.. Human gingival mesenchymal stem cell-derived exosomes cross-regulate the Wnt/β-catenin and NF-κB signalling pathways in the periodontal inflammation microenvironment. J. Clin. Periodontol., 2023, 50: 796-806

[91]

ZengT, et al.. Cartilaginous extracellular matrix enriched with human gingival mesenchymal stem cells derived “matrix bound extracellular vesicles” enabled functional reconstruction of tracheal defect. Adv. Sci., 2022, 9: 2102735

[92]

LiuZ, et al.. Local transplantation of GMSC-derived exosomes to promote vascularized diabetic wound healing by regulating the Wnt/β-catenin pathways. Nanoscale Adv., 2023, 5: 916-926

[93]

Della RoccaY, et al.. Protective effect of oral stem cells extracellular vesicles on cardiomyocytes in hypoxia-reperfusion. Front. Cell Dev. Biol., 2024, 11: 1260019

[94]

SunJ, et al.. Exosomes derived from human gingival mesenchymal stem cells attenuate the inflammatory response in periodontal ligament stem cells. Front. Chem., 2022, 10: 863364

[95]

ShiQ, et al.. GMSC-derived exosomes combined with a chitosan/silk hydrogel sponge accelerates wound healing in a diabetic rat skin defect model. Front. Physiol., 2017, 8: 904

[96]

SilvestroS, et al.. Extracellular vesicles derived from human gingival mesenchymal stem cells: a transcriptomic analysis. Genes, 2020, 11: 118

[97]

Della RoccaY, et al.. Autologous hGMSC-derived iPS: a new proposal for tissue regeneration. Int. J. Mol. Sci., 2024, 25: 9169

[98]

TianX, et al.. Gingival mesenchymal stem cell-derived exosomes are immunosuppressive in preventing collagen-induced arthritis. J. Cell. Mol. Med., 2022, 26: 693-708

[99]

WangR, et al.. Role of gingival mesenchymal stem cell exosomes in macrophage polarization under inflammatory conditions. Int. Immunopharmacol., 2020, 81: 106030

[100]

ZhangY, et al.. Effect of gingival mesenchymal stem cell-derived exosomes on inflammatory macrophages in a high-lipid microenvironment. Int. Immunopharmacol., 2021, 94: 107455

[101]

Chen, J. et al. miRNA-148a–containing GMSC-derived EVs modulate Treg/Th17 balance via IKKB/NF-κB pathway and treat a rheumatoid arthritis model. JCI Insight 9 (2024).

[102]

BrucknerS, et al.. The therapeutic effects of gingival mesenchymal stem cells and their exosomes in a chimeric model of rheumatoid arthritis. Arthritis Res. Ther., 2023, 25: 211

[103]

DiomedeF, et al.. Three-dimensional printed PLA scaffold and human gingival stem cell-derived extracellular vesicles: a new tool for bone defect repair. Stem Cell Res. Ther., 2018, 9: 1-21

[104]

RaoF, et al.. Exosomes from human gingiva-derived mesenchymal stem cells combined with biodegradable chitin conduits promote rat sciatic nerve regeneration. Stem Cells Int., 2019, 2019: 2546367

[105]

ZhangY, et al.. SIS-ECM laden with GMSC-derived exosomes promote taste bud regeneration. J. Dent. Res., 2019, 98: 225-233

[106]

WangS, et al.. Extracellular vesicles secreted by human gingival mesenchymal stem cells promote bone regeneration in rat femoral bone defects. Front. Bioeng. Biotechnol., 2023, 11: 1098172

[107]

Della RoccaY, et al.. Role of extra-cellular vesicles derived by human gingival mesenchymal stem cells in cardiomyocytes acute hypoxia. Ital. J. Anat. Embryol., 2022, 126: 35-38

[108]

GiulianiA, et al.. Could the enrichment of a biomaterial with conditioned medium or extracellular vesicles modify bone-remodeling kinetics during a defect healing? Evaluations on rat calvaria with synchrotron-based microtomography. Appl. Sci., 2020, 10: 2336

[109]

WangM, LiJ, YeY, ChenD, SongJ. SHED-derived exosomes improve the repair capacity and osteogenesis potential of hPDLCs. Oral. Dis., 2023, 29: 1692-1705

[110]

GaoY, et al.. Bioinspired porous microspheres for sustained hypoxic exosomes release and vascularized bone regeneration. Bioact. Mater., 2022, 14: 377-388

[111]

LiuF, et al.. Preliminary study on the mechanism by which exosomes derived from human exfoliated deciduous teeth improve the proliferation and osteogenic inhibitory effect of glucocorticoid-induced BMSCs. Gene, 2024, 923: 148575

[112]

ZhengY, et al.. Effects of sEV derived from SHED and DPSC on the proliferation, migration and osteogenesis of PDLSC. Regenerative Ther., 2023, 24: 489-498

[113]

FallahA, ColagarAH, KhosraviA, SaeidiM. Exosomes from SHED-MSC regulate polarization and stress oxidative indexes in THP-1 derived M1 macrophages. Arch. Biochem. Biophys., 2024, 755: 109987

[114]

JinS, et al.. Young exosome bio-nanoparticles restore aging-impaired tendon stem/progenitor cell function and reparative capacity. Adv. Mater., 2023, 35: 2211602

[115]

LiY, et al.. Engineering antioxidant poly (citrate-gallic acid)-Exosome hybrid hydrogel with microglia immunoregulation for Traumatic Brain Injury-post neuro-restoration. Compos. B: Eng., 2022, 242: 110034

[116]

BastidasJG, et al.. Secretome of stem cells from human exfoliated deciduous teeth (SHED) and its extracellular vesicles improves keratinocytes migration, viability, and attenuation of H2O2-induced cytotoxicity. Wound Repair Regeneration, 2023, 31: 827-841

[117]

Yu, T., Mi, N., Song, Y. & Xie, W. Exosomes miR-92a-3p from human exfoliated deciduous teeth inhibits periodontitis progression via the KLF4/PI3K/AKT pathway. J. Periodontal Res. 59, 771–782 (2024).

[118]

WuJ, et al.. Exosomes secreted by stem cells from human exfoliated deciduous teeth promote alveolar bone defect repair through the regulation of angiogenesis and osteogenesis. ACS Biomater. Sci. Eng., 2019, 5: 3561-3571

[119]

Jing, Y. et al. Apoptotic vesicles modulate endothelial metabolism and ameliorate ischemic retinopathy via PD1/PDL1 Axis. Adv. Healthc. Mater. 13, e2303527 (2024).

[120]

GuoJ, et al.. Exosome-shuttled mitochondrial transcription factor A mRNA promotes the osteogenesis of dental pulp stem cells through mitochondrial oxidative phosphorylation activation. Cell Prolif., 2022, 55: e13324

[121]

SunartvanichkulT, et al.. Stem cell-derived exosomes from human exfoliated deciduous teeth promote angiogenesis in hyperglycemic-induced human umbilical vein endothelial cells. J. Appl. Oral. Sci., 2023, 31: e20220427

[122]

DuZ, et al.. SHED-derived exosomes ameliorate hyposalivation caused by Sjögren’s syndrome via Akt/GSK-3β/Slug-mediated ZO-1 expression. Chin. Med. J., 2023, 136: 2596-2608

[123]

KatahiraY, et al.. Protective effects of conditioned media of immortalized stem cells from human exfoliated deciduous teeth on pressure ulcer formation. Front. Immunol., 2023, 13: 1010700

[124]

JonavičėU, et al.. Extracellular vesicles from human teeth stem cells trigger ATP release and promote migration of human microglia through P2X4 receptor/MFG-E8-dependent mechanisms. Int. J. Mol. Sci., 2021, 22: 10970

[125]

LinC-Y, et al.. The Exosomes of Stem Cells from Human Exfoliated Deciduous Teeth Suppress Inflammation in Osteoarthritis. Int. J. Mol. Sci., 2024, 25: 8560

[126]

KogaS, HoriguchiY. Efficacy of a cultured conditioned medium of exfoliated deciduous dental pulp stem cells in erectile dysfunction patients. J. Cell. Mol. Med., 2022, 26: 195-201

[127]

JonavičėU, TunaitisV, KriaučiūnaitėK, JarmalavičiūtėA, PivoriūnasA. Extracellular vesicles can act as a potent immunomodulators of human microglial cells. J. Tissue Eng. Regenerative Med., 2019, 13: 309-318

[128]

WangM, LiJ, YeY, HeS, SongJ. SHED-derived conditioned exosomes enhance the osteogenic differentiation of PDLSCs via Wnt and BMP signaling in vitro. Differentiation, 2020, 111: 1-11

[129]

JarmalavičiūtėA, TunaitisV, PivoraitėU, VenalisA, PivoriūnasA. Exosomes from dental pulp stem cells rescue human dopaminergic neurons from 6-hydroxy-dopamine–induced apoptosis. Cytotherapy, 2015, 17: 932-939

[130]

SonodaS, et al.. Targeting of deciduous tooth pulp stem cell–derived extracellular vesicles on telomerase-mediated stem cell niche and immune regulation in systemic lupus erythematosus. J. Immunol., 2021, 206: 3053-3063

[131]

ChuWX, et al.. SHED-exos promote saliva secretion by suppressing p-ERK1/2-mediated apoptosis in glandular cells. Oral. Dis., 2024, 30: 3066-3080

[132]

WeiJ, et al.. Exosomes derived from human exfoliated deciduous teeth ameliorate adult bone loss in mice through promoting osteogenesis. J. Mol. Histol., 2020, 51: 455-466

[133]

PivoraitėU, et al.. Exosomes from human dental pulp stem cells suppress carrageenan-induced acute inflammation in mice. Inflammation, 2015, 38: 1933-1941

[134]

XieY, et al.. SHED-derived exosomes promote LPS-induced wound healing with less itching by stimulating macrophage autophagy. J. Nanobiotechnol., 2022, 20 ArticleID: 239

[135]

GuoR, et al.. SHED-derived exosomes attenuate trigeminal neuralgia after CCI of the infraorbital nerve in mice via the miR-24-3p/IL-1R1/p-p38 MAPK pathway. J. Nanobiotechnol., 2023, 21 ArticleID: 458

[136]

LuH, et al.. Functional extracellular vesicles from SHEDs combined with gelatin methacryloyl promote the odontogenic differentiation of DPSCs for pulp regeneration. J. Nanobiotechnol., 2024, 22 ArticleID: 265

[137]

PengY, et al.. Young small extracellular vesicles rejuvenate replicative senescence by remodeling Drp1 translocation-mediated mitochondrial dynamics. J. Nanobiotechnol., 2024, 22 ArticleID: 543

[138]

LiY, et al.. Exosomes secreted by stem cells from human exfoliated deciduous teeth contribute to functional recovery after traumatic brain injury by shifting microglia M1/M2 polarization in rats. Stem Cell Res. Ther., 2017, 8: 1-11

[139]

LuoP, JiangC, JiP, WangM, XuJ. Exosomes of stem cells from human exfoliated deciduous teeth as an anti-inflammatory agent in temporomandibular joint chondrocytes via miR-100-5p/mTOR. Stem Cell Res. Ther., 2019, 10: 1-12

[140]

SonodaS, et al.. Extracellular vesicles from deciduous pulp stem cells recover bone loss by regulating telomerase activity in an osteoporosis mouse model. Stem Cell Res. Ther., 2020, 11: 1-16

[141]

NarbuteK, et al.. Intranasal administration of extracellular vesicles derived from human teeth stem cells improves motor symptoms and normalizes tyrosine hydroxylase expression in the substantia nigra and striatum of the 6-hydroxydopamine-treated rats. Stem Cells Transl. Med., 2019, 8: 490-499

[142]

LuoL, AverySJ, WaddingtonRJ. Exploring a chemotactic role for EVs from progenitor cell populations of human exfoliated deciduous teeth for promoting migration of naïve BMSCs in bone repair process. Stem Cells Int., 2021, 2021: 6681771

[143]

WuM, et al.. SHED aggregate exosomes shuttled miR-26a promote angiogenesis in pulp regeneration via TGF-β/SMAD2/3 signalling. Cell Prolif., 2021, 54: e13074

[144]

Huang, X. et al. Odontogenesis-empowered extracellular vesicles safeguard donor-recipient stem cell interplay to support tooth regeneration. Small 20, e2400260 (2024).

[145]

LiY, et al.. Dental stem cell-derived extracellular vesicles transfer miR-330-5p to treat traumatic brain injury by regulating microglia polarization. Int. J. oral. Sci., 2022, 14: 44

[146]

LeiF, et al.. Treatment of inflammatory bone loss in periodontitis by stem cell-derived exosomes. Acta Biomater., 2022, 141: 333-343

[147]

HuangH-M, et al.. Mechanical force-promoted osteoclastic differentiation via periodontal ligament stem cell exosomal protein ANXA3. Stem Cell Rep., 2022, 17: 1842-1858

[148]

Diomede, F. et al. A novel role in skeletal segment regeneration of extracellular vesicles released from periodontal-ligament stem cells. Int. J. Nanomed. 13, 3805–3825 (2018).

[149]

ZhengX, et al.. Biological characteristics of microRNAs secreted by exosomes of periodontal ligament stem cells due to mechanical force. Eur. J. Orthod., 2023, 45: 408-417

[150]

LanQ, et al.. Curcumin-primed periodontal ligament stem cells-derived extracellular vesicles improve osteogenic ability through the Wnt/β-catenin pathway. Front. Cell Dev. Biol., 2023, 11: 1225449

[151]

ChiricostaL, et al.. Extracellular vesicles of human periodontal ligament stem cells contain MicroRNAs associated to proto-oncogenes: Implications in cytokinesis. Front. Genet., 2020, 11: 582

[152]

Niu, Q. et al. FoxO1-overexpressed small extracellular vesicles derived from hPDLSCs promote periodontal tissue regeneration by reducing mitochondrial dysfunction to regulate osteogenesis and inflammation. Int. J. Nanomed. 19, 8751–8768 (2024).

[153]

PizzicannellaJ, et al.. Engineered extracellular vesicles from human periodontal-ligament stem cells increase VEGF/VEGFR2 expression during bone regeneration. Front. Physiol., 2019, 10: 512

[154]

KangH, LeeM-J, ParkSJ, LeeM-S. Lipopolysaccharide-preconditioned periodontal ligament stem cells induce M1 polarization of macrophages through extracellular vesicles. Int. J. Mol. Sci., 2018, 19: 3843

[155]

WangJ, et al.. Osteogenic differentiation effect of human periodontal ligament stem-cell initial cell density on autologous cells and human bone marrow stromal cells. Int. J. Mol. Sci., 2023, 24: 7133

[156]

ZhangZ, et al.. PDLSCs regulate angiogenesis of periodontal ligaments via VEGF transferred by exosomes in periodontitis. Int. J. Med. Sci., 2020, 17: 558

[157]

Zhao, B. et al. Periodontal ligament stem cell-derived small extracellular vesicles embedded in matrigel enhance bone repair through the adenosine receptor signaling pathway. Int. J. Nanomed. 17, 519-536.

[158]

Lu, J., Yu, N., Liu, Q., Xie, Y. & Zhen, L. Periodontal ligament stem cell exosomes key to regulate periodontal regeneration by miR-31-5p in mice model. Int. J. Nanomed. 18, 5327–5342 (2023).

[159]

ChangM, ChenQ, WangB, ZhangZ, HanG. Exosomes from tension force-applied periodontal ligament cells promote mesenchymal stem cell recruitment by altering microRNA profiles. Int. J. Stem Cells, 2023, 16: 202-214

[160]

KangL, MiaoY, JinY, ShenS, LinX. Exosomal miR-205-5p derived from periodontal ligament stem cells attenuates the inflammation of chronic periodontitis via targeting XBP1. Immunity. Inflamm. Dis., 2023, 11: e743

[161]

HanP, et al.. Effects of periodontal cells-derived extracellular vesicles on mesenchymal stromal cell function. J. Periodontal Res., 2023, 58: 1188-1200

[162]

ZhengY, et al.. Exosomal microRNA-155-5p from PDLSCs regulated Th17/Treg balance by targeting sirtuin-1 in chronic periodontitis. J. Cell. Physiol., 2019, 234: 20662-20674

[163]

WuY, et al.. Exosomes from cyclic stretched periodontal ligament cells induced periodontal inflammation through miR-9-5p/SIRT1/NF-κB signaling pathway. J. Immunol., 2023, 210: 2001-2015

[164]

DaiZ, et al.. Gallic acid ameliorates the inflammatory state of periodontal ligament stem cells and promotes pro-osteodifferentiation capabilities of inflammatory stem cell-derived exosomes. Life, 2022, 12: 1392

[165]

NovelloS, Tricot-DoleuxS, NovellaA, Pellen-MussiP, JeanneS. Influence of periodontal ligament stem cell-derived conditioned medium on osteoblasts. Pharmaceutics, 2022, 14: 729

[166]

ZhaoY, et al.. The experimental study of periodontal ligament stem cells derived exosomes with hydrogel accelerating bone regeneration on alveolar bone defect. Pharmaceutics, 2022, 14: 2189

[167]

CuiS, et al.. Small extracellular vesicles from periodontal ligament stem cells primed by lipopolysaccharide regulate macrophage M1 polarization via miR-433-3p targeting TLR2/TLR4/NF-κB. Inflammation, 2023, 46: 1849-1858

[168]

PourhajibagherM, BahadorA. Periodontal ligament stem cell-derived exosome-loaded Emodin mediated antimicrobial photodynamic therapy against cariogenic bacteria. BMC Oral. Health, 2024, 24 ArticleID: 311

[169]

LuY, et al.. Rab27a-mediated extracellular vesicle secretion contributes to osteogenesis in periodontal ligament-bone niche communication. Sci. Rep., 2023, 13 ArticleID: 8479

[170]

Soundara RajanT, et al.. Human periodontal ligament stem cells secretome from multiple sclerosis patients suppresses NALP3 inflammasome activation in experimental autoimmune encephalomyelitis. Int. J. Immunopathol. Pharmacol., 2017, 30: 238-252

[171]

RajanTS, et al.. The secretome of periodontal ligament stem cells from MS patients protects against EAE. Sci. Rep., 2016, 6 ArticleID: 38743

[172]

XuX-Y, et al.. Exosomes derived from P2X7 receptor gene-modified cells rescue inflammation-compromised periodontal ligament stem cells from dysfunction. Stem Cells Transl. Med., 2020, 9: 1414-1430

[173]

LiuT, et al.. Human periodontal ligament stem cell-derived exosomes promote bone regeneration by altering MicroRNA profiles. Stem Cells Int., 2020, 2020: 8852307

[174]

LiuM, et al.. Exosomal miR-141-3p from PDLSCs alleviates high glucose-induced senescence of PDLSCs by activating the KEAP1-NRF2 signaling pathway. Stem Cells Int., 2023, 2023: 7136819

[175]

HanP, et al.. 3D bioprinted small extracellular vesicles from periodontal cells enhance mesenchymal stromal cell function. Biomater. Adv., 2024, 158: 213770

[176]

YuJ, et al.. Effect of psoralen on the regulation of osteogenic differentiation induced by periodontal stem cell-derived exosomes. Hum. Cell, 2023, 36: 1389-1402

[177]

XiangM, et al.. Metformin enhances the therapeutic effects of extracellular vesicles derived from human periodontal ligament stem cells on periodontitis. Sci. Rep., 2024, 14 ArticleID: 19940

[178]

DongJ, et al.. Dental pulp stem cell-derived small extracellular vesicle in irradiation-induced senescence. Biochem. Biophys. Res. Commun., 2021, 575: 28-35

[179]

ChansaenrojA, et al.. Magnetic bioassembly platforms towards the generation of extracellular vesicles from human salivary gland functional organoids for epithelial repair. Bioact. Mater., 2022, 18: 151-163

[180]

TianJ, et al.. Small extracellular vesicles derived from hypoxic preconditioned dental pulp stem cells ameliorate inflammatory osteolysis by modulating macrophage polarization and osteoclastogenesis. Bioact. Mater., 2023, 22: 326-342

[181]

ShenZ, et al.. Chitosan hydrogel incorporated with dental pulp stem cell-derived exosomes alleviates periodontitis in mice via a macrophage-dependent mechanism. Bioact. Mater., 2020, 5: 1113-1126

[182]

ZengJ, et al.. Exosomes from human umbilical cord mesenchymal stem cells and human dental pulp stem cells ameliorate lipopolysaccharide-induced inflammation in human dental pulp stem cells. Arch. Oral. Biol., 2022, 138: 105411

[183]

LiB, et al.. Hypoxia preconditioned DPSC-derived exosomes regulate angiogenesis via transferring LOXL2. Exp. Cell Res., 2023, 425: 113543

[184]

VakhshitehF, et al.. Exosomes derived from miR-34a-overexpressing mesenchymal stem cells inhibit in vitro tumor growth: A new approach for drug delivery. Life Sci., 2021, 266: 118871

[185]

ZhangS, et al.. Human dental pulp stem cell-derived exosomes decorated titanium scaffolds for promoting bone regeneration. Colloids Surf. B: Biointerfaces, 2024, 235: 113775

[186]

ChenW-J, et al.. The role of small extracellular vesicles derived from lipopolysaccharide-preconditioned human dental pulp stem cells in dental pulp regeneration. J. Endod., 2021, 47: 961-969

[187]

HanS, et al.. Programmed release of vascular endothelial growth factor and exosome from injectable chitosan nanofibrous microsphere-based PLGA-PEG-PLGA hydrogel for enhanced bone regeneration. Int. J. Biol. Macromol., 2023, 253: 126721

[188]

HuangC-C, NarayananR, AlapatiS, RavindranS. Exosomes as biomimetic tools for stem cell differentiation: applications in dental pulp tissue regeneration. Biomaterials, 2016, 111: 103-115

[189]

GuoH, et al.. Odontogenesis-related developmental microenvironment facilitates deciduous dental pulp stem cell aggregates to revitalize an avulsed tooth. Biomaterials, 2021, 279: 121223

[190]

SwansonWB, et al.. Controlled release of odontogenic exosomes from a biodegradable vehicle mediates dentinogenesis as a novel biomimetic pulp capping therapy. J. Control. Rel., 2020, 324: 679-694

[191]

KlimovaD, et al.. Extracellular vesicles derived from dental mesenchymal stem/stromal cells with gemcitabine as a cargo have an inhibitory effect on the growth of pancreatic carcinoma cell lines in vitro. Mol. Cell. Probes, 2023, 67: 101894

[192]

OgataK, et al.. Extracellular vesicles of iPS cells highly capable of producing HGF and TGF-β1 can attenuate Sjögren’s syndrome via innate immunity regulation. Cell. Signal., 2024, 113: 110980

[193]

WinkelA, et al.. Cell culture media notably influence properties of human mesenchymal stroma/stem-like cells from different tissues. Cytotherapy, 2020, 22: 653-668

[194]

LiZ, et al.. Apoptotic vesicles activate autophagy in recipient cells to induce angiogenesis and dental pulp regeneration. Mol. Ther., 2022, 30: 3193-3208

[195]

SwansonWB, et al.. Scaffolds with controlled release of pro-mineralization exosomes to promote craniofacial bone healing without cell transplantation. Acta Biomater., 2020, 118: 215-232

[196]

Lin, T.-Y. et al. 2, 3, 5, 4′-tetrahydroxystilbene-2-O-β-D-glucoside–stimulated dental pulp stem cells-derived exosomes for wound healing and bone regeneration. J. Dental Sci. 20, 154–163 (2024).

[197]

Mas-BarguesC, et al.. Small extracellular vesicles from senescent stem cells trigger adaptive mechanisms in young stem cells by increasing antioxidant enzyme expression. Redox Biol., 2023, 62: 102668

[198]

AltanerovaU, et al.. Dental pulp mesenchymal stem/stromal cells labeled with iron sucrose release exosomes and cells applied intra-nasally migrate to intracerebral glioblastoma. Neoplasma, 2016, 63: 925-933

[199]

NakatsukaR, SasakiY, MasutaniM, NozakiT. PARP1 regulates cellular processes mediated by exosomal miRNAs in dental pulp stem cells. J. Hard Tissue Biol., 2021, 30: 371-378

[200]

AbdelgawadLM, NghnughiMH, AbdelgwadM. Influence of photo biomodulation using 980 nm diode laser and exsosomes derived from dental pulp stem cells on pulp regeneration of dogs’ teeth. Reactions, 2022, 9: 11

[201]

VenugopalC, et al.. by human dental pulp mesenchymal stem cells: from billions to nano. Curr. Gene Ther., 2018, 18: 307-323

[202]

LiY, et al.. Deciphering the heterogeneity landscape of mesenchymal stem/stromal cell-derived extracellular vesicles for precise selection in translational medicine. Adv. Healthc. Mater., 2023, 12: 2202453

[203]

Fei, Y., Ling, Z., Tong, Q.& Wang, J. Apoptotic extracellular vesicles from supernumerary tooth-derived pulp stem cells transfer COL1A1 to promote angiogenesis via PI3K/Akt/VEGF pathway. Int. J. Nanomed. 19, 6811–6828 (2024).

[204]

DiomedeF, et al.. Extracellular vesicles (EVs): a promising therapeutic tool in the heart tissue regeneration. BioFactors, 2024, 50: 509-522

[205]

XieL, et al.. Exosomal circLPAR1 promoted osteogenic differentiation of homotypic dental pulp stem cells by competitively binding to hsa-miR-31. Biomed. Res. Int., 2020, 2020: 6319395

[206]

Mas-BarguesC, et al.. Extracellular vesicles from healthy cells improves cell function and stemness in premature senescent stem cells by miR-302b and HIF-1α activation. Biomolecules, 2020, 10: 957

[207]

LiB, et al.. Hypoxia alters the proteome profile and enhances the angiogenic potential of dental pulp stem cell-derived exosomes. Biomolecules, 2022, 12: 575

[208]

LiS, et al.. Dental pulp stem cell-derived exosomes alleviate cerebral ischaemia-reperfusion injury through suppressing inflammatory response. Cell Prolif., 2021, 54: e13093

[209]

ZhouH, et al.. Periodontitis-compromised dental pulp stem cells secrete extracellular vesicles carrying miRNA-378a promote local angiogenesis by targeting Sufu to activate the Hedgehog/Gli1 signalling. Cell Prolif., 2021, 54: e13026

[210]

MerckxG, et al.. Angiogenic effects of human dental pulp and bone marrow-derived mesenchymal stromal cells and their extracellular vesicles. Cells, 2020, 9: 312

[211]

ChaiY, et al.. Dental pulp stem cell-derived exosomes promote sciatic nerve regeneration via optimizing schwann cell function. Cell. Reprogramming, 2024, 26: 67-78

[212]

WangS, et al.. Fabrication of an exosome-loaded thermosensitive chitin-based hydrogel for dental pulp regeneration. J. Mater. Chem. B, 2023, 11: 1580-1590

[213]

LiJ, JuY, LiuS, FuY, ZhaoS. Exosomes derived from lipopolysaccharide-preconditioned human dental pulp stem cells regulate Schwann cell migration and differentiation. Connect. Tissue Res., 2021, 62: 277-286

[214]

FaruquFN, et al.. Three-dimensional culture of dental pulp pluripotent-like stem cells (DPPSCs) enhances Nanog expression and provides a serum-free condition for exosome isolation. FASEB BioAdvances, 2020, 2: 419

[215]

Sánchez-SánchezR, et al.. miR-4732-3p in extracellular vesicles from mesenchymal stromal cells is cardioprotective during myocardial ischemia. Front. Cell Dev. Biol., 2021, 9: 734143

[216]

UmarS, et al.. Immunomodulatory properties of naïve and inflammation-informed dental pulp stem cell derived extracellular vesicles. Front. Immunol., 2024, 15: 1447536

[217]

GuoS, et al.. Stimulating extracellular vesicles production from engineered tissues by mechanical forces. Nano Lett., 2021, 21: 2497-2504

[218]

Altanerova, U. et al. Human mesenchymal stem cell-derived iron oxide exosomes allow targeted ablation of tumor cells via magnetic hyperthermia. Int. J. Nanomed. 12, 7923–7936 (2017).

[219]

LinT, et al.. Inhibition of chondrocyte apoptosis in a rat model of osteoarthritis by exosomes derived from miR-140-5p-overexpressing human dental pulp stem cells. Int. J. Mol. Med., 2021, 47: 1-

[220]

ZhouZ, et al.. Exosomes derived from dental pulp stem cells accelerate cutaneous wound healing by enhancing angiogenesis via the Cdc42/p38 MAPK pathway. Int. J. Mol. Med., 2022, 50: 1-15

[221]

ZhangS, et al.. Extracellular vesicles-loaded fibrin gel supports rapid neovascularization for dental pulp regeneration. Int. J. Mol. Sci., 2020, 21: 4226

[222]

Gómez-FerrerM, et al.. HIF-1α and pro-inflammatory signaling improves the immunomodulatory activity of MSC-derived extracellular vesicles. Int. J. Mol. Sci., 2021, 22: 3416

[223]

GaneshV, et al.. Exosome-based cell homing and angiogenic differentiation for dental pulp regeneration. Int. J. Mol. Sci., 2022, 24: 466

[224]

FuY, CuiS, ZhouY, QiuL. Dental pulp stem cell-derived exosomes alleviate mice knee osteoarthritis by inhibiting TRPV4-mediated osteoclast activation. Int. J. Mol. Sci., 2023, 24: 4926

[225]

Teixeira, M. R., Alievi, A. L., da Costa, V. R., Kerkis, I. & Araldi, R. P. Exploring the therapeutic potential of extracellular vesicles derived from human immature dental pulp cells on papillary thyroid cancer. Int. J. Mol. Sci. 25, 8178 (2024).

[226]

Chen, Y. et al. The application of pulp tissue derived-exosomes in pulp regeneration: a novel cell-homing approach. Int. J. Nanomed. 17, 465-476 (2022).

[227]

IvicaA, GhayorC, ZehnderM, ValdecS, WeberFE. Pulp-derived exosomes in a fibrin-based regenerative root filling material. J. Clin. Med., 2020, 9: 491

[228]

JinQ, et al.. Extracellular vesicles derived from human dental pulp stem cells promote osteogenesis of adipose-derived stem cells via the MAPK pathway. J. Tissue Eng., 2020, 11: 2041731420975569

[229]

TerunumaA, et al.. Extracellular vesicles from mesenchymal stem cells of dental pulp and adipose tissue display distinct transcriptomic characteristics suggestive of potential therapeutic targets. J. Stem Cells Regenerative Med., 2021, 17: 56

[230]

LeeA, et al.. DPSC-derived extracellular vesicles promote rat jawbone regeneration. J. Dent. Res., 2023, 102: 313-321

[231]

Eren BelginE, GençD, TekinL, SezginS, AladağA. Anti-Inflammatory effect of dental pulpa mesenchymal stem cell exosomes loaded mucoadhesive hydrogel on mice with dental nickel hypersensitivity. Macromol. Biosci., 2024, 24: 2300352

[232]

LiL, GeJ. Exosome-derived lncRNA-Ankrd26 promotes dental pulp restoration by regulating miR-150-TLR4 signaling. Mol. Med. Rep., 2022, 25: 1-11

[233]

NasiriK. Exosomes derived from human dental stem cell enhance the viability of odontoblasts. Nanomed. Res. J., 2023, 8: 365-372

[234]

KongF, et al.. Dental pulp stem cell-derived extracellular vesicles mitigate haematopoietic damage after radiation. Stem Cell Rev. Rep., 2021, 17: 318-331

[235]

JiL, et al.. Comparison of immunomodulatory properties of exosomes derived from bone marrow mesenchymal stem cells and dental pulp stem cells. Immunologic Res., 2019, 67: 432-442

[236]

Chai, Y. et al. Study on the role and mechanism of exosomes derived from dental pulp stem cells in promoting regeneration of myelin sheath in rats with sciatic nerve injury. Mol. Neurobiol. 61, 6175–6188 (2024).

[237]

LiuC, et al.. Dental pulp stem cell-derived exosomes suppress M1 macrophage polarization through the ROS-MAPK-NFκB P65 signaling pathway after spinal cord injury. J. Nanobiotechnol., 2022, 20 ArticleID: 65

[238]

LiangX, et al.. Dental pulp mesenchymal stem cell-derived exosomes inhibit neuroinflammation and microglial pyroptosis in subarachnoid hemorrhage via the miRNA-197-3p/FOXO3 axis. J. Nanobiotechnol., 2024, 22 ArticleID: 426

[239]

HuS, et al.. Dental pulp stem cell-derived exosomes revitalize salivary gland epithelial cell function in NOD mice via the GPER-mediated cAMP/PKA/CREB signaling pathway. J. Transl. Med., 2023, 21 ArticleID: 361

[240]

HuX, et al.. Lineage-specific exosomes promote the odontogenic differentiation of human dental pulp stem cells (DPSCs) through TGFβ1/smads signaling pathway via transfer of microRNAs. Stem Cell Res. Ther., 2019, 10: 1-14

[241]

ZhouH, et al.. The proangiogenic effects of extracellular vesicles secreted by dental pulp stem cells derived from periodontally compromised teeth. Stem cell Res. Ther., 2020, 11: 1-18

[242]

ZhengJ, et al.. MicroRNA-enriched small extracellular vesicles possess odonto-immunomodulatory properties for modulating the immune response of macrophages and promoting odontogenesis. Stem Cell Res. Ther., 2020, 11: 1-14

[243]

LuoX, et al.. Odontoblasts release exosomes to regulate the odontoblastic differentiation of dental pulp stem cells. Stem Cell Res. Ther., 2023, 14: 176

[244]

ChiY, LiuT, JinQ, LiuH. Extracellular vesicles carrying RUNX3 promote differentiation of dental pulp stem cells. Tissue Eng. Regenerative Med., 2024, 21: 111-122

[245]

ImanishiY, et al.. Efficacy of extracellular vesicles from dental pulp stem cells for bone regeneration in rat calvarial bone defects. Inflamm. Regeneration, 2021, 41: 1-10

[246]

Amaro-PrellezoE, et al.. Extracellular vesicles from dental pulp mesenchymal stem cells modulate macrophage phenotype during acute and chronic cardiac inflammation in athymic nude rats with myocardial infarction. Inflamm. Regeneration, 2024, 44 ArticleID: 25

[247]

HeitzerM, et al.. Evaluation of in vitro biocompatibility of human pulp stem cells with allogeneic, alloplastic, and xenogeneic grafts under the influence of extracellular vesicles. Sci. Rep., 2023, 13 ArticleID: 12475

[248]

ShimizuY, et al.. Exosomes from dental pulp cells attenuate bone loss in mouse experimental periodontitis. J. Periodontal Res., 2022, 57: 162-172

[249]

MiaoY, et al.. Transfer of miR-877–3p via extracellular vesicles derived from dental pulp stem cells attenuates neuronal apoptosis and facilitates early neurological functional recovery after cerebral ischemia–reperfusion injury through the Bclaf1/P53 signaling pathway. Pharmacol. Res., 2024, 206: 107266

[250]

ChenT-Y, et al.. Exosomes derived from Polygonum multiflorum-treated human dental pulp stem cells (hDPSCs): New approach in regenerative medicine. J. Drug Deliv. Sci. Technol., 2024, 99: 105941

[251]

DiomedeF, et al.. Decellularized dental pulp, extracellular vesicles, and 5-azacytidine: a new tool for endodontic regeneration. Biomedicines, 2022, 10: 403

[252]

HeX, et al.. MSC-derived exosome promotes M2 polarization and enhances cutaneous wound healing. Stem Cells Int., 2019, 2019: 7132708

[253]

LiX, et al.. Exosomes derived from maxillary BMSCs enhanced the osteogenesis in iliac BMSCs. Oral. Dis., 2020, 26: 131-144

[254]

ZhuQ, et al.. Exosomes derived from mesenchymal stromal cells promote bone regeneration by delivering miR-182–5p-inhibitor. Pharmacol. Res., 2023, 192: 106798

[255]

XuS, WangZ. Bone marrow mesenchymal stem cell-derived exosomes enhance osteoclastogenesis during alveolar bone deterioration in rats. RSC Adv., 2017, 7: 21153-21163

[256]

LamparskiHG, et al.. Production and characterization of clinical grade exosomes derived from dendritic cells. J. Immunol. Methods, 2002, 270: 211-226

[257]

WebberJ, ClaytonA. How pure are your vesicles?. J. Extracell. Vesicles, 2013, 2: 19861

[258]

Momen-HeraviF, et al.. Current methods for the isolation of extracellular vesicles. Biol. Chem., 2013, 394: 1253-1262

[259]

HanP, LaiA, SalomonC, IvanovskiS. Detection of salivary small extracellular vesicles associated inflammatory cytokines gene methylation in gingivitis. Int. J. Mol. Sci., 2020, 21: 5273

[260]

KimJY, et al.. Defined MSC exosome with high yield and purity to improve regenerative activity. J. Tissue Eng., 2021, 12: 20417314211008626

[261]

FarshidfarN, et al.. The feasible application of microfluidic tissue/organ-on-a-chip as an impersonator of oral tissues and organs: a direction for future research. Bio-Des. Manuf., 2023, 6: 478-506

[262]

FaruquFN, et al.. Three-dimensional culture of dental pulp pluripotent-like stem cells (DPPSCs) enhances Nanog expression and provides a serum-free condition for exosome isolation. FASEB bioAdvances, 2020, 2: 419-433

[263]

PatelGK, et al.. Comparative analysis of exosome isolation methods using culture supernatant for optimum yield, purity and downstream applications. Sci. Rep., 2019, 9 ArticleID: 5335

[264]

HeM, ZengY. Microfluidic exosome analysis toward liquid biopsy for cancer. J. Lab. Autom., 2016, 21: 599-608

[265]

WangJ, MaP, KimDH, LiuB-F, DemirciU. Towards microfluidic-based exosome isolation and detection for tumor therapy. Nano Today, 2021, 37: 101066

[266]

ZhaoZ, YangY, ZengY, HeM. A microfluidic ExoSearch chip for multiplexed exosome detection towards blood-based ovarian cancer diagnosis. Lab Chip, 2016, 16: 489-496

[267]

Hassanpour TamrinS, Sanati NezhadA, SenA. Label-free isolation of exosomes using microfluidic technologies. ACS Nano, 2021, 15: 17047-17079

[268]

YakubovichE, PolischoukA, EvtushenkoV. Principles and problems of exosome isolation from biological fluids. Biochem. (Mosc.), Suppl. Ser. A: Membr. Cell Biol., 2022, 16: 115-126

[269]

LiuDS, et al.. Size-exclusion chromatography as a technique for the investigation of novel extracellular vesicles in cancer. Cancers, 2020, 12: 3156

[270]

GuoJ, et al.. Establishment of a simplified dichotomic size-exclusion chromatography for isolating extracellular vesicles toward clinical applications. J. Extracell. Vesicles, 2021, 10: e12145

[271]

KangYT, et al.. Isolation and profiling of circulating tumor-associated exosomes using extracellular vesicular lipid–protein binding affinity based microfluidic device. Small, 2019, 15: 1903600

[272]

FarhanaFZ, et al.. Isolation and detection of exosomes using Fe2O3 nanoparticles. ACS Appl. Nano Mater., 2021, 4: 1175-1186

[273]

RayamajhiS, AryalS. Surface functionalization strategies of extracellular vesicles. J. Mater. Chem. B., 2020, 8: 4552-4569

[274]

GonzálezMI, et al.. Covalently labeled fluorescent exosomes for in vitro and in vivo applications. Biomedicines, 2021, 9: 81

[275]

YamamotoM, et al.. Application of high-mannose-type glycan-specific lectin from Oscillatoria Agardhii for affinity isolation of tumor-derived extracellular vesicles. Anal. Biochem., 2019, 580: 21-29

[276]

KangY-T, et al.. High-purity capture and release of circulating exosomes using an exosome-specific dual-patterned immunofiltration (ExoDIF) device. Nanoscale, 2017, 9: 13495-13505

[277]

IliukA, et al.. Plasma-derived extracellular vesicle phosphoproteomics through chemical affinity purification. J. Proteome Res., 2020, 19: 2563-2574

[278]

LouD, WangY, YangQ, HuL, ZhuQ. Ultrafiltration combing with phospholipid affinity-based isolation for metabolomic profiling of urinary extracellular vesicles. J. Chromatogr. A, 2021, 1640: 461942

[279]

TayebiM, ZhouY, TripathiP, ChandramohanadasR, AiY. Exosome purification and analysis using a facile microfluidic hydrodynamic trapping device. Anal. Chem., 2020, 92: 10733-10742

[280]

KimJ, LeeH, ParkK, ShinS. Rapid and efficient isolation of exosomes by clustering and scattering. J. Clin. Med., 2020, 9: 650

[281]

VisanKS, et al.. Comparative analysis of tangential flow filtration and ultracentrifugation, both combined with subsequent size exclusion chromatography, for the isolation of small extracellular vesicles. J. Extracell. Vesicles, 2022, 11: 12266

[282]

RamnauthN, et al.. Development of a microfluidic device for exosome isolation in point-of-care settings. Sensors, 2023, 23: 8292

[283]

LigaA, VliegenthartA, OosthuyzenW, DearJ, Kersaudy-KerhoasM. Exosome isolation: a microfluidic road-map. Lab Chip, 2015, 15: 2388-2394

[284]

SatoYT, et al.. Engineering hybrid exosomes by membrane fusion with liposomes. Sci. Rep., 2016, 6 ArticleID: 21933

[285]

WuY, et al.. Microfluidic technology for the isolation and analysis of exosomes. Micromachines, 2022, 13: 1571

[286]

YongT, et al.. Tumor exosome-based nanoparticles are efficient drug carriers for chemotherapy. Nat. Commun., 2019, 10 ArticleID: 3838

[287]

HuT, et al.. Flow-electricity coupling fields enhance microfluidic platforms for efficient exosome isolation. Anal. Methods, 2024, 16: 5335-5344

[288]

SaniF, et al.. Unveiling exosomes: Cutting-edge isolation techniques and their therapeutic potential. J. Cell. Mol. Med., 2024, 28: e70139

[289]

JiaY, et al.. Small extracellular vesicles isolation and separation: Current techniques, pending questions and clinical applications. Theranostics, 2022, 12: 6548

[290]

KimD, et al.. Ultra-thin membrane filter with a uniformly arrayed nanopore structure for nanoscale separation of extracellular vesicles without cake formation. Nanoscale Adv., 2023, 5: 640-649

[291]

AliakbariF, et al.. A methodological primer of extracellular vesicles isolation and characterization via different techniques. Biol. Methods Protoc., 2024, 9: bpae009

[292]

ShiL, et al.. Rapid and label-free isolation of small extracellular vesicles from biofluids utilizing a novel insulator based dielectrophoretic device. Lab Chip, 2019, 19: 3726-3734

[293]

GaoJ, et al.. Recent developments in isolating methods for exosomes. Front. Bioeng. Biotechnol., 2023, 10: 1100892

[294]

BrennanK, et al.. A comparison of methods for the isolation and separation of extracellular vesicles from protein and lipid particles in human serum. Sci. Rep., 2020, 10 ArticleID: 1039

[295]

DouY, RenL, KulabhusanPK, ZaripovEA, BerezovskiMV. Quantitative capillary electrophoresis for analysis of extracellular vesicles (EVqCE). Separations, 2021, 8: 110

[296]

YangD, et al.. Progress, opportunity, and perspective on exosome isolation-efforts for efficient exosome-based theranostics. Theranostics, 2020, 10: 3684

[297]

BányaiA, et al.. Dean-flow affected lateral focusing and separation of particles and cells in periodically inhomogeneous microfluidic channels. Sensors, 2023, 23: 800

[298]

PourariaH, FoudaziR, HoustonJP. Exploitation of elasto-inertial fluid flow for the separation of nano-sized particles: Simulating the isolation of extracellular vesicles. Cytometry A, 2023, 103: 786-795

[299]

ShiriF, et al.. Separation of U87 glioblastoma cell-derived small and medium extracellular vesicles using elasto-inertial flow focusing (a spiral channel). Sci. Rep., 2022, 12 ArticleID: 6146

[300]

BhagatAAS, KuntaegowdanahalliSS, PapautskyI. Continuous particle separation in spiral microchannels using dean flows and differential migration. Lab Chip, 2008, 8: 1906-1914

[301]

PatelK, StarkH. Instability of a liquid sheet with viscosity contrast in inertial microfluidics. Eur. Phys. J. E, 2021, 44 ArticleID: 144

[302]

ZhangJ, et al.. Fundamentals and applications of inertial microfluidics: a review. Lab a Chip, 2016, 16: 10-34

[303]

ShiR. Numerical simulation of inertial microfluidics: a review. Eng. Appl. Comput. Fluid Mech., 2023, 17: 2177350

[304]

NamJ, JangWS, HongDH, LimCS. Viscoelastic separation and concentration of fungi from blood for highly sensitive molecular diagnostics. Sci. Rep., 2019, 9 ArticleID: 3067

[305]

LiuC, et al.. Field-free isolation of exosomes from extracellular vesicles by microfluidic viscoelastic flows. ACS Nano, 2017, 11: 6968-6976

[306]

HettiarachchiS, et al.. Viscoelastic microfluidics for enhanced separation resolution of submicron particles and extracellular vesicles. Nanoscale, 2024, 16: 3560-3570

[307]

Mukherjee, S. & Sarkar, K. Lateral migration of a viscoelastic drop in a Newtonian fluid in a shear flow near a wall. Phys. Fluids 26 (2014).

[308]

SevenlerD, TonerM. High throughput intracellular delivery by viscoelastic mechanoporation. Nat. Commun., 2024, 15 ArticleID: 115

[309]

KumarT, et al.. High throughput viscoelastic particle focusing and separation in spiral microchannels. Sci. Rep., 2021, 11 ArticleID: 8467

[310]

Jiang D., Ni C., Tang W., Huang D., Xiang N. Inertial microfluidics in contraction–expansion microchannels: a review. Biomicrofluidics 15 (2021).

[311]

CollinsDJ, AlanT, NeildA. Particle separation using virtual deterministic lateral displacement (vDLD). Lab Chip, 2014, 14: 1595-1603

[312]

TottoriN, NisisakoT. Tunable deterministic lateral displacement of particles flowing through thermo-responsive hydrogel micropillar arrays. Sci. Rep., 2023, 13 ArticleID: 4994

[313]

Bowman T. J., Drazer G., Frechette J. Inertia and scaling in deterministic lateral displacement. Biomicrofluidics 7 (2013).

[314]

ZhangH, LydenD. Asymmetric-flow field-flow fractionation technology for exomere and small extracellular vesicle separation and characterization. Nat. Protoc., 2019, 14: 1027-1053

[315]

ChiangC, YehJ, YehC. Nanoparticles separation by different conditions at asymmetric flow field-flow fractionation. J. Mech., 2023, 39: 500-507

[316]

ShendrukTN, et al.. Field-flow fractionation and hydrodynamic chromatography on a microfluidic chip. Anal. Chem., 2013, 85: 5981-5988

[317]

PetersenKE, et al.. A review of exosome separation techniques and characterization of B16-F10 mouse melanoma exosomes with AF4-UV-MALS-DLS-TEM. Anal. Bioanal. Chem., 2014, 406: 7855-7866

[318]

KimYB, LeeGB, MoonMH. Size separation of exosomes and microvesicles using flow field-flow fractionation/multiangle light scattering and lipidomic comparison. Anal. Chem., 2022, 94: 8958-8965

[319]

PriedolsM, et al.. Bifurcated Asymmetric Field Flow Fractionation of Nanoparticles in PDMS-Free Microfluidic Devices for Applications in Label-Free Extracellular Vesicle Separation. Polymers, 2023, 15: 789

[320]

ChronakisMI, Von Der AuM, MeermannB. Single cell-asymmetrical flow field-flow fractionation/ICP-time of flight-mass spectrometry (sc-AF4/ICP-ToF-MS): an efficient alternative for the cleaning and multielemental analysis of individual cells. J. Anal. Spectrom., 2022, 37: 2691-2700

[321]

DrexelR, et al.. Fast and purification-free characterization of bio-nanoparticles in biological media by electrical asymmetrical flow field-flow fractionation hyphenated with multi-angle light scattering and nanoparticle tracking analysis detection. Molecules, 2020, 25: 4703

[322]

MitranoD, NeubauerK, RanvilleJ, ThomasR. Field-flow fractionation coupled with ICP-MS forthe analysis of engineered nanoparticles in environmental samples. Spectroscopy, 2012, 25: 652-664

[323]

LuX, XuanX. Elasto-inertial pinched flow fractionation for continuous shape-based particle separation. Anal. Chem., 2015, 87: 11523-11530

[324]

Chiriacò, M. S. et al. Lab-on-chip for exosomes and microvesicles detection and characterization. Sensors 18, 3175 (2018).

[325]

ShinS, et al.. Separation of extracellular nanovesicles and apoptotic bodies from cancer cell culture broth using tunable microfluidic systems. Sci. Rep., 2017, 7: 1-8

[326]

Sunahiro, S., Senaha, M., Yamada, M. & Seki, M., editors. Pinched Flow Fractionization device for sizes and density dependent separation of particles utilizing centrifugal pumping. In: 12th International Conference on Miniaturized Systems for Chemistry and Life Sciences, San Diego, California, USA (2008).

[327]

LiuF, et al.. IB-LBM study on cell sorting by pinched flow fractionation. Bio-Med. Mater. Eng., 2014, 24: 2547-2554

[328]

HickeySM, et al.. Fluorescence microscopy—an outline of hardware, biological handling, and fluorophore considerations. Cells, 2021, 11: 35

[329]

KimA, NgWB, BerntW, ChoN-J. Validation of size estimation of nanoparticle tracking analysis on polydisperse macromolecule assembly. Sci. Rep., 2019, 9 ArticleID: 2639

[330]

JainA, PosnerJD. Particle dispersion and separation resolution of pinched flow fractionation. Anal. Chem., 2008, 80: 1641-1648

[331]

RisbudSR, DrazerG. Mechanism governing separation in microfluidic pinched flow fractionation devices. Microfluidics Nanofluidics, 2014, 17: 1003-1009

[332]

WangZ, et al.. Acoustofluidic separation enables early diagnosis of traumatic brain injury based on circulating exosomes. Microsyst. Nanoeng., 2021, 7: 20

[333]

FanY, WangX, RenJ, LinF, WuJ. Recent advances in acoustofluidic separation technology in biology. Microsyst. Nanoeng., 2022, 8: 94

[334]

NaquinTD, et al.. Acoustic separation and concentration of exosomes for nucleotide detection: ASCENDx. Sci. Adv., 2024, 10: eadm8597

[335]

WuM, et al.. Acoustofluidic separation of cells and particles. Microsyst. Nanoeng., 2019, 5: 32

[336]

TayebiM, YangD, CollinsDJ, AiY. Deterministic sorting of submicrometer particles and extracellular vesicles using a combined electric and acoustic field. Nano Lett., 2021, 21: 6835-6842

[337]

ZhangJ, et al.. A solution to the biophysical fractionation of extracellular vesicles: acoustic nanoscale separation via wave-pillar excitation resonance (ANSWER). Sci. Adv., 2022, 8: eade0640

[338]

Díaz-Reinoso, B. Concentration and Purification of Seaweed Extracts Using Membrane Technologies. Sustainable Seaweed Technologies 371–390 (Elsevier, 2020).

[339]

LiangL-G, et al.. An integrated double-filtration microfluidic device for isolation, enrichment and quantification of urinary extracellular vesicles for detection of bladder cancer. Sci. Rep., 2017, 7 ArticleID: 46224

[340]

WooH-K, et al.. Exodisc for rapid, size-selective, and efficient isolation and analysis of nanoscale extracellular vesicles from biological samples. ACS Nano, 2017, 11: 1360-1370

[341]

LiuF, et al.. The exosome total isolation chip. ACS Nano, 2017, 11: 10712-10723

[342]

WangZ, et al.. Ciliated micropillars for the microfluidic-based isolation of nanoscale lipid vesicles. Lab Chip, 2013, 13: 2879-2882

[343]

YasuiT, et al.. Unveiling massive numbers of cancer-related urinary-microRNA candidates via nanowires. Sci. Adv., 2017, 3: e1701133

[344]

HanZ, et al.. Highly efficient exosome purification from human plasma by tangential flow filtration based microfluidic chip. Sens. Actuators B: Chem., 2021, 333: 129563

[345]

LewisJM, et al.. Integrated analysis of exosomal protein biomarkers on alternating current electrokinetic chips enables rapid detection of pancreatic cancer in patient blood. ACS Nano, 2018, 12: 3311-3320

[346]

ShiL, RanaA, EsfandiariL. A low voltage nanopipette dielectrophoretic device for rapid entrapment of nanoparticles and exosomes extracted from plasma of healthy donors. Sci. Rep., 2018, 8: 1-12

[347]

DaviesRT, et al.. Microfluidic filtration system to isolate extracellular vesicles from blood. Lab Chip, 2012, 12: 5202-5210

[348]

MogiK, HayashidaK, YamamotoT. Damage-less handling of exosomes using an ion-depletion zone in a microchannel. Anal. Sci., 2018, 34: 875-880

[349]

MarczakS, et al.. Simultaneous isolation and preconcentration of exosomes by ion concentration polarization. Electrophoresis, 2018, 39: 2029-2038

[350]

ChoS, et al.. Isolation of extracellular vesicle from blood plasma using electrophoretic migration through porous membrane. Sens. Actuators B: Chem., 2016, 233: 289-297

[351]

TayHM, et al.. Rapid purification of sub-micrometer particles for enhanced drug release and microvesicles isolation. NPG Asia Mater., 2017, 9, ArticleID: e434-e

[352]

ZhouY, MaZ, TayebiM, AiY. Submicron particle focusing and exosome sorting by wavy microchannel structures within viscoelastic fluids. Anal. Chem., 2019, 91: 4577-4584

[353]

WunschBH, et al.. Nanoscale lateral displacement arrays for the separation of exosomes and colloids down to 20 nm. Nat. Nanotechnol., 2016, 11: 936-940

[354]

SantanaSM, AntonyakMA, CerioneRA, KirbyBJ. Microfluidic isolation of cancer-cell-derived microvesicles from hetergeneous extracellular shed vesicle populations. Biomed. Microdevices, 2014, 16: 869-877

[355]

SmithJT, et al.. Integrated nanoscale deterministic lateral displacement arrays for separation of extracellular vesicles from clinically-relevant volumes of biological samples. Lab Chip, 2018, 18: 3913-3925

[356]

PetersenKE, et al.. Exosome isolation: cyclical electrical field flow fractionation in low-ionic-strength fluids. Anal. Chem., 2018, 90: 12783-12790

[357]

ShinS, et al.. Separation of extracellular nanovesicles and apoptotic bodies from cancer cell culture broth using tunable microfluidic systems. Sci. Rep., 2017, 7 ArticleID: 9907

[358]

LeeK, ShaoH, WeisslederR, LeeH. Acoustic purification of extracellular microvesicles. ACS nNano, 2015, 9: 2321-2327

[359]

EvanderM, GidlöfO, OldeB, ErlingeD, LaurellT. Non-contact acoustic capture of microparticles from small plasma volumes. Lab Chip, 2015, 15: 2588-2596

[360]

WuM, et al.. Isolation of exosomes from whole blood by integrating acoustics and microfluidics. Proc. Natl. Acad. Sci., 2017, 114: 10584-10589

[361]

Yeo, J. C. et al. Label-free extraction of extracellular vesicles using centrifugal microfluidics. Biomicrofluidics 12 (2018).

[362]

SidhomK, ObiPO, SaleemA. A review of exosomal isolation methods: is size exclusion chromatography the best option?. Int. J. Mol. Sci., 2020, 21: 6466

[363]

Zhang, Y. et al. Exosome: a review of its classification, isolation techniques, storage, diagnostic and targeted therapy applications. Int. J. Nanomed. 15, 6917-6934 (2020).

[364]

YangX-X, SunC, WangL, GuoX-L. New insight into isolation, identification techniques and medical applications of exosomes. J. Control. Rel., 2019, 308: 119-129

[365]

ThéryC, AmigorenaS, RaposoG, ClaytonA. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr. Protoc. Cell Biol., 2006, 30: 3.22. 1-3. 9

[366]

LiuW-Z, MaZ-J, KangX-W. Current status and outlook of advances in exosome isolation. Anal. Bioanal. Chem., 2022, 414: 7123-7141

[367]

HeL, et al.. Ovarian cancer cell-secreted exosomal miR-205 promotes metastasis by inducing angiogenesis. Theranostics, 2019, 9: 8206

[368]

LivshitsMA, et al.. Isolation of exosomes by differential centrifugation: Theoretical analysis of a commonly used protocol. Sci. Rep., 2015, 5: 1-14

[369]

LinH, et al.. Advances of exosomes in periodontitis treatment. J. Transl. Med., 2022, 20 ArticleID: 279

[370]

WengY, et al.. Effective isolation of exosomes with polyethylene glycol from cell culture supernatant for in-depth proteome profiling. Analyst, 2016, 141: 4640-4646

[371]

RiderMA, HurwitzSN, MeckesDG. ExtraPEG: a polyethylene glycol-based method for enrichment of extracellular vesicles. Sci. Rep., 2016, 6: 1-14

[372]

LudwigA-K, et al.. Precipitation with polyethylene glycol followed by washing and pelleting by ultracentrifugation enriches extracellular vesicles from tissue culture supernatants in small and large scales. J. Extracell. Vesicles, 2018, 7: 1528109

[373]

WitwerKW, et al.. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J. Extracell. Vesicles, 2013, 2: 20360

[374]

ZhouH, et al.. Collection, storage, preservation, and normalization of human urinary exosomes for biomarker discovery. Kidney Int., 2006, 69: 1471-1476

[375]

OosthuyzenW, et al.. Quantification of human urinary exosomes by nanoparticle tracking analysis. J. Physiol., 2013, 591: 5833-5842

RIGHTS & PERMISSIONS

The Author(s)

AI Summary AI Mindmap
PDF

144

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/