AccScience Publishing / IJB / Volume 9 / Issue 6 / DOI: 10.36922/ijb.0114
Cite this article
359
Download
2167
Views
Journal Browser
Volume | Year
Issue
Search
News and Announcements
View All
REVIEW

Exosome-based bioinks for 3D bioprinting applications in tissue engineering and regenerative medicine

Qian Wang1 Yang Liu1 Shuqing Zhang1 Fan He2 Tong Shi3 Jizong Li3* Zhimin Wang4* Jia Jia1,2*
Show Less
1 School of Life Sciences, Shanghai University, Shanghai, China
2 Sino-Swiss Institute of Advanced Technology, School of Micro-electronics, Shanghai University, Shanghai, China
3 Shanghai Center of Biomedicine Development, Shanghai, China
4 Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Shanghai, China
Submitted: 4 April 2023 | Accepted: 12 May 2023 | Published: 7 July 2023
(This article belongs to the Special Issue 3D printing for tissue engineering and regenerative medicine)
© 2023 by the Author(s). This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

Bioprinting is an emerging technology for tissue engineering and regenerative medicine. Despite its fast, accurate manufacture for tissues and organs in vitro, bioprinting has been seriously limited for biofabrication because of the restricted approaches to reproducing the extracellular matrix (ECM) with sufficient bioactivities for bioprinted cells. Exosomes are natural biological particles with proteins, lipids, or genetic materials. They have distinct properties and unique biological functions to manipulate cellular behaviors and cell fates, showing great potential to support cells for bioprinting. Here, we reviewed the recent progresses of exosome-advanced bioprinting for tissue engineering and regenerative medicine. Firstly, we offer an overview of the basics of exosomes and the current representative applications of exosomes in bone tissue engineering, immunological regulations, angiogenesis, and neural regenerations. Then, a brief introduction about the bioinks and the currently developed bioprinting methods is provided. We further give an in-depth review of the biomedical applications of bioprinting with exosomes, majorly in bone engineering, vascular engineering, therapy of neuron injury, and skin regeneration. We also conclude with an outlook on the challenges of the unmet needs of bioprinting cells with correspondent ECM environments through bioprinting with exosomes.

Keywords
Exosomes
Bioprinting
Bioinks
Regenerative medicine
Tissue engineering
Funding
Zhimin Wang reports financial support was provided by National Natural Science Foundation of China (No. 51833006) and Innovation Promotion Program of NHC and Shanghai Key Labs, SIBPT (No. CX2023-03). Jia Jia reports financial support was provided by Shanghai University (No. N-58-D305-22-204). Jizong Li reports financial support was provided by Science and Technology Commission of Shanghai Municipality (No. 17DZ2260600 & No. 22692116403).
References
  1. Matai I, Kaur G, Seyedsalehi A, et al., 2020, Progress in 3D bioprinting technology for tissue/organ regenerative engineering. Biomaterials, 226: 119536. https://doi.org/10.1016/j.biomaterials.2019.119536

 

 

  1. Heinrich MA, et al., 2019, 3D bioprinting: From benches to translational applications. Small, 15: e1805510. https://doi.org/10.1002/smll.201805510

 

 

  1. Tavafoghi M, Darabi MA, Mahmoodi M, et al., 2021, Multimaterial bioprinting and combination of processing techniques towards the fabrication of biomimetic tissues and organs. Biofabrication, 13(4): 042002. https://doi.org/10.1088/1758-5090/ac0b9a

 

 

  1. Murphy SV, Atala A, 2014, 3D bioprinting of tissues and organs. Nat Biotechnol, 32: 773–785. https://doi.org/10.1038/nbt.2958

 

 

  1. Samandari M, Quint J, Rodríguez-delaRosa A, et al., 2022, Bioinks and bioprinting strategies for skeletal muscle tissue engineering. Adv Mater, 34: e2105883. https://doi.org/10.1002/adma.202105883

 

 

  1. Dobos A, Gantner F, Markovic M, et al., 2021, On-chip high-definition bioprinting of microvascular structures. Biofabrication, 13: 015016. https://doi.org/10.1088/1758-5090/abb063

 

 

  1. Freeman FE, Burdis R, Kelly DJ, 2021, Printing new bones: From print-and-implant devices to bioprinted bone organ precursors. Trends Mol Med, 27: 700–711. https://doi.org/10.1016/j.molmed.2021.05.001

 

 

  1. Zhou F, Hong Y, Liang R, et al., 2020, Rapid printing of bio-inspired 3D tissue constructs for skin regeneration. Biomaterials, 258: 120287. https://doi.org/10.1016/j.biomaterials.2020.120287

 

 

  1. Moghaddam AS, Khonakdar, HA, Arjmand M, et al., 2021, Review of bioprinting in regenerative medicine: Naturally derived bioinks and stem cells. ACS Appl Biomater, 4: 4049– 4070. https://doi.org/10.1021/acsabm.1c00219

 

 

  1. Herrmann IK, Wood MJA, Fuhrmann G, 2021, Extracellular vesicles as a next-generation drug delivery platform. Nat Nanotechnol, 16: 748–759. https://doi.org/10.1038/s41565-021-00931-2

 

 

  1. van Niel G, D’Angelo G, Raposo G, 2018, Shedding light on the cell biology of extracellular vesicles. Nat Rev, 19: 213–228. https://doi.org/10.1038/nrm.2017.125

 

 

  1. Pan BT, Johnstone RM, 1983, Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the receptor. Cell, 33: 967–978. https://doi.org/10.1016/0092-8674(83)90040-5

 

 

  1. Harding C, Heuser J, Stahl P, 1983, Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J Cell Biol, 97: 329–339. https://doi.org/10.1083/jcb.97.2.329

 

 

  1. Pan BT, Blostein R, Johnstone RM, 1983, Loss of the transferrin receptor during the maturation of sheep reticulocytes in vitro. An immunological approach. Biochem J, 210: 37–47. https://doi.org/10.1042/bj2100037

 

 

  1. Valadi H, Ekström K, Bossios, et al., 2007, Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol, 9: 654–659. https://doi.org/10.1038/ncb1596

 

 

  1. Katsuda T, Kosaka N, Takeshita F, et al., 2013, The therapeutic potential of mesenchymal stem cell-derived extracellular vesicles. Proteomics, 13: 1637–1653. https://doi.org/10.1002/pmic.201200373

 

 

  1. Yáñez-Mó M, Siljander PRM, Andreu Z, et al., 2015, Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles, 4(1): 27066. https://doi.org/10.3402/jev.v4.27066

 

 

  1. Kalluri R, LeBleu VS, 2020, The biology, function, and biomedical applications of exosomes. Science, 367: eaau6977

 

 

  1. Budnik V, Ruiz-Cañada C, Wendler F, 2016, Extracellular vesicles round off communication in the nervous system. Nat Rev, 17: 160–172. https://doi.org/10.1038/nrn.2015.29

 

 

  1. Zhang S, Chuah SJ, Lai RC, et al., 2018, MSC exosomes mediate cartilage repair by enhancing proliferation, attenuating apoptosis and modulating immune reactivity. Biomaterials, 156: 16–27. https://doi.org/10.1016/j.biomaterials.2017.11.028

 

 

  1. Kim YG, Choi J, Kim K, 2020, Mesenchymal stem cell-derived exosomes for effective cartilage tissue repair and treatment of osteoarthritis. Biotechnol J, 15: e2000082. https://doi.org/10.1002/biot.202000082

 

 

  1. Jiang S, Tian G, Yang Z, et al., 2021, Enhancement of acellular cartilage matrix scaffold by Wharton’s jelly mesenchymal stem cell-derived exosomes to promote osteochondral regeneration. Bioact Mater, 6: 2711–2728. https://doi.org/10.1016/j.bioactmat.2021.01.031

 

 

  1. Hongxing Hu, Lanlan Dong, Ziheng Bu, et al., 2020, miR- 23a-3p-abundant small extracellular vesicles released from Gelma/nanoclay hydrogel for cartilage regeneration. J Extracell Vesicles, 9: 1778883. https://doi.org/10.1080/20013078.2020.1778883

 

 

  1. Wang, W, Liang X, Zheng K, et al., 2022, Horizon of exosome-mediated bone tissue regeneration: The all-rounder role in biomaterial engineering. Mater Today Bio, 16: 100355. https://doi.org/10.1016/j.mtbio.2022.100355

 

 

  1. Wang G, Xie L, Li B, et al., 2021, A nanounit strategy reverses immune suppression of exosomal PD-L1 and is associated with enhanced ferroptosis. Nat Commun, 12: 5733. https://doi.org/10.1038/s41467-021-25990-w

 

 

  1. Ding G, Zhou L, Qian Y, et al., 2015, Pancreatic cancer-derived exosomes transfer miRNAs to dendritic cells and inhibit RFXAP expression via miR-212-3p. Oncotarget, 6: 29877–29888. https://doi.org/10.18632/oncotarget.4924

 

 

  1. Huang P, Wang L, Li Q, et al., 2020, Atorvastatin enhances the therapeutic efficacy of mesenchymal stem cells-derived exosomes in acute myocardial infarction via up-regulating long non-coding RNA H19. Cardiovasc Res, 116: 353–367. https://doi.org/10.1093/cvr/cvz139

 

 

  1. Ma T, Chen Y, Chen Y, et al., 2018, MicroRNA-132, Delivered by mesenchymal stem cell-derived exosomes, promote angiogenesis in myocardial infarction. Stem Cell Int, 2018: 3290372. https://doi.org/10.1155/2018/3290372

 

 

  1. Andaloussi SEL, Mäger I, Breakefield XO, et al., 2013, Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat Rev, 12: 347–357. https://doi.org/10.1038/nrd3978

 

 

  1. Haney MJ, Klyachko NL, Zhao Y, et al., 2015, Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J Control Release, 207: 18–30. https://doi.org/10.1016/j.jconrel.2015.03.033

 

 

  1. Lykke-Andersen S, Brodersen DE, Jensen TH, 2009, Origins and activities of the eukaryotic exosome. J Cell Sci, 122: 1487–1494. https://doi.org/10.1242/jcs.047399

 

 

  1. Garreta E, Oria R, Tarantino C, et al., 2017, Tissue engineering by decellularization and 3D bioprinting. Mater Today, 20: 166–178. https://doi.org/10.1016/j.mattod.2016.12.005

 

 

  1. Santschi M, Vernengo A, Eglin D, et al., 2019, Decellularized matrix as a building block in bioprinting and electrospinning. Curr Opin BiomedEng, 10: 116–122. https://doi.org/10.1016/j.cobme.2019.05.003

 

 

  1. Yang D, Zhang W, Zhang H, et al., 2020, Progress, opportunity, and perspective on exosome isolation - efforts for efficient exosome-based theranostics. Theranostics, 10: 3684–3707. https://doi.org/10.7150/thno.41580

 

 

  1. Gu C, Feng J, Waqas A, et al., 2021, Technological advances of 3D scaffold-based stem cell/exosome therapy in tissues and organs. Front Cell Dev Biol, 9: 709204. https://doi.org/10.3389/fcell.2021.709204

 

 

  1. Sun YH, Zhang BJ, Zhai D, et al., 2021, Three-dimensional printing of bioceramic-induced macrophage exosomes: Immunomodulation and osteogenesis/angiogenesis. Npg Asia Mater, 13(1): 72. https://doi.org/10.1038/s41427-021-00340-w

 

 

  1. Tang Q, Lu B, He J, et al., 2022, Exosomes-loaded thermosensitive hydrogels for corneal epithelium and stroma regeneration. Biomaterials, 280: 121320. https://doi.org/10.1016/j.biomaterials.2021.121320

 

 

  1. Lin J, Wang Z, Huang J, et al., 2021, Microenvironment-protected exosome-hydrogel for facilitating endometrial regeneration, fertility restoration, and live birth of offspring. Small, 17(11): 2007235. https://doi.org/10.1002/smll.202007235

 

 

  1. Rolland TJ, Peterson TE, Singh RD, et al., 2022, Exosome biopotentiated hydrogel restores damaged skeletal muscle in a porcine model of stress urinary incontinence (vol 29, 58, 2022). NPJ Regen Medi, 7(1): 1–17. https://doi.org/10.1038/s41536-022-00260-5

 

 

  1. Zhang Y, Huo M, Wang Y, et al., 2022, A tailored bioactive 3D porous poly(lactic-acid)-exosome scaffold with osteo-immunomodulatory and osteogenic differentiation properties. J BiolEng, 16(1):1–14. https://doi.org/10.1186/s13036-022-00301-z

 

 

  1. Li Q, Gong S, Yao W, et al., 2021, Exosome loaded genipin crosslinked hydrogel facilitates full thickness cutaneous wound healing in rat animal model. Drug Deliv, 28: 884–893, https://doi.org/10.1080/10717544.2021.1912210

 

 

  1. Fan L, Liu C, Chen X, et al., 2022, Exosomes-loaded electroconductive hydrogel synergistically promotes tissue repair after spinal cord injury via immunoregulation and enhancement of myelinated axon growth. Adv Sci, 9(13): 2105586. https://doi.org/10.1002/advs.202105586

 

 

  1. Liu S, Li R, Dou K, et al., 2023, Injectable thermo-sensitive hydrogel containing ADSC-derived exosomes for the treatment of cavernous nerve injury. Carbohydr Polym, 300: 120226. https://doi.org/10.1016/j.carbpol.2022.120226

 

 

  1. Li Y, Wang Min, Sun Meng, et al., 2022, Engineering antioxidant poly (citrate-gallic acid)-exosome hybrid hydrogel with microglia immunoregulation for traumatic brain injury-post neuro-restoration. Compos Part B-Eng, 242: 110034. https://doi.org/10.1016/j.compositesb.2022.110034

 

 

  1. Yu Z, Hao R, Du J, et al., 2022, A human cornea-on-a-chip for the study of epithelial wound healing by extracellular vesicles. iScience, 25: 104200. https://doi.org/10.1016/j.isci.2022.104200

 

 

  1. Shafei S, Khanmohammadi M, Heidari R, et al., 2020, Exosome loaded alginate hydrogel promotes tissue regeneration in full-thickness skin wounds: An in vivo study. J Biomed Mater Res Part A, 108: 545–556. https://doi.org/10.1002/jbm.a.36835

 

 

  1. Badylak SF, Taylor D, Uygun K, 2011, Annual Review of Biomedical Engineering, Annual Reviews Inc, 27–53.

 

 

  1. Song JJ, Ott HC, 2011, Organ engineering based on decellularized matrix scaffolds. Trends Mol Med, 17: 424–432. https://doi.org/10.1016/j.molmed.2011.03.005

 

 

  1. Londono R, Badylak SF, 2015, Biologic scaffolds for regenerative medicine: Mechanisms of in vivo remodeling. Ann Biomed Eng, 43: 577–592. https://doi.org/10.1007/s10439-014-1103-8

 

 

  1. Yi HG, Jeong YH, Kim Y, et al., 2019, A bioprinted human-glioblastoma-on-a-chip for the identification of patient-specific responses to chemoradiotherapy. Nat Biomedi Eng, 3: 509–519. https://doi.org/10.1038/s41551-019-0363-x

 

 

  1. Wu Y, Wang J, Shi Y, et al., 2017, Implantation of brain-derived extracellular matrix enhances neurological recovery after traumatic brain injury. Cell Transplant, 26: 1224–1234. https://doi.org/10.1177/0963689717714090

 

 

  1. Barrs RW, Jia J, Silver SE, et al., 2020, Biomaterials for bioprinting microvasculature. Chem Rev, 120: 10887–10949. https://doi.org/10.1021/acs.chemrev.0c00027

 

 

  1. Valot L, Martinez J, Mehdi A, et al., 2019, Chemical insights into bioinks for 3D printing. Chem Soc Rev, 48: 4049–4086. https://doi.org/10.1039/c7cs00718c

 

 

  1. Ozbolat IT, Hospodiuk M, 2016, Current advances and future perspectives in extrusion-based bioprinting. Biomaterials, 76: 321–343. https://doi.org/10.1016/j.biomaterials.2015.10.076

 

 

  1. Xia Z, Jin S, Ye K, 2018, Tissue and organ 3D bioprinting. SLAS Technol, 23: 301–314. https://doi.org/10.1177/2472630318760515

 

 

  1. Leucht A, Volz AC, Rogal J, et al., 2020, Advanced gelatin-based vascularization bioinks for extrusion-based bioprinting of vascularized bone equivalents. Sci Rep, 10: 5330. https://doi.org/10.1038/s41598-020-62166-w

 

 

  1. Li X, Liu B, Pei B, et al., 2020, Inkjet bioprinting of biomaterials. Chem Rev, 120: 10793–10833. https://doi.org/10.1021/acs.chemrev.0c00008

 

 

  1. Miri AK, Khalilpour A, Cecen B, et al., 2019, Multiscale bioprinting of vascularized models. Biomaterials, 198: 204– 216. https://doi.org/10.1016/j.biomaterials.2018.08.006

 

 

  1. Suntornnond R, Ng WL, Huang X, et al., 2022, Improving printability of hydrogel-based bio-inks for thermal inkjet bioprinting applications via saponification and heat treatment processes. J Mater Chem B, 10: 5989–6000. https://doi.org/10.1039/d2tb00442a

 

 

  1. Malda J, Visser J, Melchels FP, et al., 2013, 25th anniversary article: Engineering hydrogels for biofabrication. Adv Mater, 25: 5011–5028. https://doi.org/10.1002/adma.201302042

 

 

  1. Ng WL, Lee JM, Zhou M, et al., 2020, Vat polymerization-based bioprinting-process, materials, applications and regulatory challenges. Biofabrication, 12: 022001. https://doi.org/10.1088/1758-5090/ab6034

 

 

  1. Zhang B, Gao L, Ma L, et al., 2019, 3D bioprinting: A novel avenue for manufacturing tissues and organs. Engineering, 5: 777–794. https://doi.org/10.1016/j.eng.2019.03.009

 

 

  1. Kingsley DM, Roberge CL, Rudkouskaya A, et al., 2019, Laser-based 3D bioprinting for spatial and size control of tumor spheroids and embryoid bodies. Acta Biomater, 95: 357–370. https://doi.org/10.1016/j.actbio.2019.02.014

 

 

  1. Sohn H-S, Oh J-K, 2019, Review of bone graft and bone substitutes with an emphasis on fracture surgeries. Biomater Res, 23(1): 1–7. https://doi.org/10.1186/s40824-019-0157-y

 

 

  1. Liang Q, Ma Y, Yao X, et al., 2022, Advanced 3D-printing bioinks for articular cartilage. Int J Bioprint, 8: 15–30. https://doi.org/10.18063/ijb.v8i3.511

 

 

  1. Fan J, Lee CS, Kim S, et al., 2020, Generation of small RNA-modulated exosome mimetics for bone regeneration. ACS Nano, 14: 11973–11984. https://doi.org/10.1021/acsnano.0c05122

 

 

  1. Zha Y, Li Y, Lin T, et al., 2021, Progenitor cell-derived exosomes endowed with VEGF plasmids enhance osteogenic induction and vascular remodeling in large segmental bone defects. Theranostics, 11: 397–409. https://doi.org/10.7150/thno.50741

 

 

  1. Chen P, Zheng L, Wang Y, et al., 2019, Desktop-stereolithography 3D printing of a radially oriented extracellular matrix/mesenchymal stem cell exosome bioink for osteochondral defect regeneration. Theranostics, 9: 2439–2459. https://doi.org/10.7150/thno.31017

 

 

  1. Wu Z, Pu P, Su Z, et al., 2020, Schwann cell-derived exosomes promote bone regeneration and repair by enhancing the biological activity of porous Ti6Al4V scaffolds. Biochem BiophysRes Commun, 531: 559–565. https://doi.org/10.1016/j.bbrc.2020.07.094

 

 

  1. Zha Y, Lin T, Li Y, et al., 2020, Exosome-mimetics as an engineered gene-activated matrix induces in-situ vascularized osteogenesis. Biomaterials, 247: 119985. https://doi.org/10.1016/j.biomaterials.2020.119985

 

 

  1. Li F, Wu J, Li D, et al., 2022, Engineering stem cells to produce exosomes with enhanced bone regeneration effects: An alternative strategy for gene therapy. J Nanobiotechnol, 20(1): 1–23. https://doi.org/10.1186/s12951-022-01347-3

 

 

  1. Zhang K, Zhao X, Chen X, et al., 2018, Enhanced therapeutic effects of mesenchymal stem cell-derived exosomes with an injectable hydrogel for hindlimb ischemia treatment. ACS Appl Mater Interfaces, 10: 30081–30091. https://doi.org/10.1021/acsami.8b08449

 

 

  1. Nagappan PG, Chen H, Wang DY, 2020, Neuroregeneration and plasticity: A review of the physiological mechanisms for achieving functional recovery postinjury. Mil Med Res, 7: 30. https://doi.org/10.1186/s40779-020-00259-3

 

 

  1. Poongodi R, Chen Y-L, Yang T-H, et al., 2021, Bio-scaffolds as cell or exosome carriers for nerve injury repair. Int J Mol Sci, 22: https://doi.org/10.3390/ijms222413347

 

 

  1. Liu X, Wang J, Wang P, et al., 2022, Hypoxia-pretreated mesenchymal stem cell-derived exosomes-loaded low-temperature extrusion 3D-printed implants for neural regeneration after traumatic brain injury in canines. Front Bioeng Biotechnol, 10: 1025138. https://doi.org/10.3389/fbioe.2022.1025138

 

 

  1. Han M, Yang H, Lu X, et al., 2022, Three-dimensional-cultured MSC-derived exosome-hydrogel hybrid microneedle array patch for spinal cord repair. Nano Lett, 22: 6391–6401. https://doi.org/10.1021/acs.nanolett.2c02259

 

 

  1. Li L, Zhang Y, Mu J, et al., 2020, Transplantation of human mesenchymal stem-cell-derived exosomes immobilized in an adhesive hydrogel for effective treatment of spinal cord injury. Nano Lett, 20: 4298–4305. https://doi.org/10.1021/acs.nanolett.0c00929

 

 

  1. Hsu JM, Shiue SJ, Yang KD, et al., Locally applied stem cell exosome-scaffold attenuates nerve injury-induced pain in rats. J Pain Res, 13: 3257–3268. https://doi.org/10.2147/JPR.S286771

 

 

  1. Rao F, Zhang D, Fang T, et al., 2019, Exosomes from human gingiva-derived mesenchymal stem cells combined with biodegradable chitin conduits promote rat sciatic nerve regeneration. Stem Cells Int, 2019: 2546367. https://doi.org/10.1155/2019/2546367

 

 

  1. Liu Z, Tong H, Li J, et al., 2022, Low-stiffness hydrogels promote peripheral nerve regeneration through the rapid release of exosomes. Front Bioeng Biotechnol, 10: 922570. https://doi.org/10.3389/fbioe.2022.922570

 

 

  1. Wang T, Li Y, Guo M, et al., 2021, Exosome-mediated delivery of the neuroprotective peptide PACAP38 promotes retinal ganglion cell survival and axon regeneration in rats with traumatic optic neuropathy. Front Cell Develop Biol, 9: 659783. https://doi.org/10.3389/fcell.2021.659783

 

 

  1. Sun BK, Siprashvili Z, Khavari PA, 2014, Advances in skin grafting and treatment of cutaneous wounds. Science, 346: 941–945. https://doi.org/10.1126/science.1253836

 

 

  1. Avci P, Gupta A, Sadasivam M, et al., 2013, Low-level laser (light) therapy (LLLT) in skin: Stimulating, healing, restoring. Semin Cutan Med Surg, 32: 41–52

 

 

  1. Xiong M, Zhang Q, Hu W, et al., 2021, The novel mechanisms and applications of exosomes in dermatology and cutaneous medical aesthetics. Pharmacol Res, 166: 105490. https://doi.org/10.1016/j.phrs.2021.105490

 

 

  1. Wang WM, Wu C, Jin HZ, 2019, Exosomes in chronic inflammatory skin diseases and skin tumors. Exp Dermatol, 28: 213–218. https://doi.org/10.1111/exd.13857

 

 

  1. Griffiths CEM, Cumberbatch M, Tucker SC, et al., 2001, Exogenous topical lactoferrin inhibits allergen-induced Langerhans cell migration and cutaneous inflammation in humans. Br J Dermatol, 144: 715–725. https://doi.org/10.1046/j.1365-2133.2001.04125.x

 

 

  1. An Y, Lin S, Tan X, et al., 2021, Exosomes from adipose-derived stem cells and application to skin wound healing. Cell Prolif, 54: e12993. https://doi.org/10.1111/cpr.12993

 

 

  1. Shi A, Li J, Qiu X, et al., 2021, TGF-β loaded exosome enhances ischemic wound healing in vitro and in vivo. Theranostics, 11: 6616–6631. https://doi.org/10.7150/thno.57701

 

 

  1. Holl J, Kowalewski C, Zimek Z, et al., 2021, Chronic diabetic wounds and their treatment with skin substitutes. Cells, 10(3): 655. https://doi.org/10.3390/cells10030655

 

 

  1. Wang M, Wang C, Chen M, et al., 2019, Efficient angiogenesis-based diabetic wound healing/skin reconstruction through bioactive antibacterial adhesive ultraviolet shielding nanodressing with exosome release. ACS Nano, 13: 10279–10293. https://doi.org/10.1021/acsnano.9b03656

 

 

  1. Huang J, Xiong J, Yang L, et al., 2021, Cell-free exosome-laden scaffolds for tissue repair. Nanoscale, 13: 8740–8750. https://doi.org/10.1039/d1nr01314a

 

 

  1. Samaeekia R, Rabiee B, Putra I, et al., 2018, Effect of human corneal mesenchymal stromal cell-derived exosomes on corneal epithelial wound healing. Invest Ophthalmol Vis Sci, 59: 5194–5200. https://doi.org/10.1167/iovs.18-24803

 

 

  1. Shojaati G, Khandaker I, Funderburgh ML, et al., 2019, Mesenchymal stem cells reduce corneal fibrosis and inflammation via extracellular vesicle-mediated delivery of miRNA. Stem Cells Transl Med, 8: 1192–1201. https://doi.org/10.1002/sctm.18-0297

 

 

  1. Sorkio A, Koch L, Koivusalo L, et al., 2018, Human stem cell based corneal tissue mimicking structures using laser-assisted 3D bioprinting and functional bioinks. Biomaterials, 171: 57–71. https://doi.org/10.1016/j.biomaterials.2018.04.034

 

 

  1. Isaacson A, Swioklo S, Connon CJ, 2018, 3D bioprinting of a corneal stroma equivalent. Exp Eye Res, 173: 188–193. https://doi.org/10.1016/j.exer.2018.05.010

 

 

  1. Ji L, Bao L, Gu Z, et al., 2019, Comparison of immunomodulatory properties of exosomes derived from bone marrow mesenchymal stem cells and dental pulp stem cells. Immunol Res, 67: 432–442. https://doi.org/10.1007/s12026-019-09088-6

 

 

  1. Wei J, Song Y, Du Z, et al., 2020, Exosomes derived from human exfoliated deciduous teeth ameliorate adult bone loss in mice through promoting osteogenesis. J Mol Histol, 51: 455–466. https://doi.org/10.1007/s10735-020-09896-3

 

 

  1. Tian Y, Liu M, Liu Y, et al., 2021, The performance of 3D bioscaffolding based on a human periodontal ligament stem cell printing technique. J Biomed Mater Res Part A, 109: 1209–1219. https://doi.org/10.1002/jbm.a.37114

 

 

  1. Gómez-Guillén MC, Giménez B, López-Caballero ME, et al., Functional and bioactive properties of collagen and gelatin from alternative sources: A review. Food Hydrocoll, 25: 1813–1827. https://doi.org/10.1016/j.foodhyd.2011.02.007

 

 

  1. Rowley JA, Madlambayan G, Mooney DJ, 1999, Alginate hydrogels as synthetic extracellular matrix materials. Biomaterials, 20: 45–53. https://doi.org/10.1016/S0142-9612(98)00107-0

 

 

  1. Norotte C, Marga FS, Niklason LE, et al., 2009, Scaffold-free vascular tissue engineering using bioprinting. Biomaterials, 30: 5910–5917. https://doi.org/10.1016/j.biomaterials.2009.06.034

 

 

  1. Billiet T, Vandenhaute M, Schelfhout J, et al., 2012, A review of trends and limitations in hydrogel-rapid prototyping for tissue engineering. Biomaterials, 33: 6020–6041. https://doi.org/10.1016/j.biomaterials.2012.04.050

 

 

  1. Samanta S, Rajasingh S, Drosos N, et al., 2018, Exosomes: New molecular targets of diseases. Acta Pharmacologica Sinica, 39: 501–513. https://doi.org/10.1038/aps.2017.162

 

 

  1. Kishore R, Khan M, 2017, Cardiac cell-derived exosomes: Changing face of regenerative biology. Eur Heart J, 38: 212–215. https://doi.org/10.1093/eurheartj/ehw324

 

 

  1. Faught E, Henrickson L, Vijayan MM, 2017, Plasma exosomes are enriched in Hsp70 and modulated by stress and cortisol in rainbow trout. J Endocrinol, 232: 237–246. https://doi.org/10.1530/JOE-16-0427

 

 

  1. Hong CS, Funk S, Muller L, et al., 2016, Isolation of biologically active and morphologically intact exosomes from plasma of patients with cancer. J Extracell Vesicles, 5: 29289. https://doi.org/10.3402/jev.v5.29289

 

 

  1. Zarovni N, Corrado A, Guazzi P, et al., 2015, Integrated isolation and quantitative analysis of exosome shuttled proteins and nucleic acids using immunocapture approaches. Methods, 87: 46–58. https://doi.org/10.1016/j.ymeth.2015.05.028

 

 

  1. Zhang Y, Liu Y, Liu H, et al., 2019, Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci, 9: 19. https://doi.org/10.1186/s13578-019-0282-2

 

 

  1. Hu Y, Li X, Zhang Q, et al., 2021, Exosome-guided bone targeted delivery of Antagomir-188 as an anabolic therapy for bone loss. Bioact Mater, 6: 2905–2913. https://doi.org/10.1016/j.bioactmat.2021.02.014

 

  1. Ma S, Wu J, Hu H, et al., 2022, Novel fusion peptides deliver exosomes to modify injectable thermo-sensitive hydrogels for bone regeneration. Mater Today Bio, 13: 100195. https://doi.org/10.1016/j.mtbio.2021.100195

 

 

  1. Zhang FX, Liu P, Ding W, et al., 2021, Injectable mussel-inspired highly adhesive hydrogel with exosomes for endogenous cell recruitment and cartilage defect regeneration. Biomaterials, 278: 121169. https://doi.org/10.1016/j.biomaterials.2021.121169

 

 

  1. Chen L, Yu C, Xiong Y, et al., 2022, Multifunctional hydrogel enhances bone regeneration through sustained release of stromal cell-derived factor-1α and exosomes. Bioact Mater, 25: 460–471. https://doi.org/10.1016/j.bioactmat.2022.07.030

 

 

  1. Zhao Y, Gong Y, Liu X, et al., 2022, The experimental study of periodontal ligament stem cells derived exosomes with hydrogel accelerating bone regeneration on alveolar bone defect. Pharmaceutics, 14(10): 2189. https://doi.org/10.3390/pharmaceutics14102189

 

 

  1. Wang L, Wang J, Zhou X, et al., 2020, A new self-healing hydrogel containing hucMSC-derived exosomes promotes bone regeneration. Front Bioeng Biotechnol, 8: 564731. https://doi.org/10.3389/fbioe.2020.564731

 

 

  1. Mu J, Li L, Wu J, et al., 2022, Hypoxia-stimulated mesenchymal stem cell-derived exosomes loaded by adhesive hydrogel for effective angiogenic treatment of spinal cord injury. Biomater Sci, 10: 1803–1811. https://doi.org/10.1039/d1bm01722e

 

 

  1. Shi Q, Qian Z, Liu D, et al., 2017, GMSC-derived exosomes combined with a chitosan/silk hydrogel sponge accelerates wound healing in a diabetic rat skin defect model. Front Physiol, 8: 904. https://doi.org/10.3389/fphys.2017.00904

 

 

  1. Han C, Zhou J, Liang C, et al., 2019, Human umbilical cord mesenchymal stem cell derived exosomes encapsulated in functional peptide hydrogels promote cardiac repair. Biomater Sci, 7(7): 2920–2933.

 

 

  1. Wang C, Wang M, Xu T, et al., 2019, Engineering bioactive self-healing antibacterial exosomes hydrogel for promoting chronic diabetic wound healing and complete skin regeneration. Theranostics, 9: 65–76. https://doi.org/10.7150/thno.29766

 

 

 



 

 

Conflict of interest
The authors declare no conflict of interest.
Share
Back to top
International Journal of Bioprinting, Electronic ISSN: 2424-8002 Print ISSN: 2424-7723, Published by AccScience Publishing