Overcoming Challenges in the Clinical Translation of MSC-Exosomes: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Subjects: Ophthalmology
Contributor: , , , , , ,

MSC-based therapy for ophthalmic diseases has limitations in biocompatibility, penetration, and delivery. To address these challenges, researchers are exploring MSC exosomes, which possess similar properties to MSCs and efficiently deliver therapeutic factors to difficult-to-target ocular tissues.

Exosomes, derived from MSCs, share the same therapeutic properties as their parent cells, including anti-inflammatory, anti-apoptotic, and immunomodulatory effects, making them a promising alternative to MSCs for ocular therapy [1-3]. Exosomes possess the advantage of nano-size, enabling them to better penetrate biological barriers such as the blood-retinal barrier and deliver their cargo effectively to ocular tissues. Additionally, their cargo is protected from degradation, increasing their bioavailability and potential for ocular drug-delivery applications.

Recent research has shown that MSC-derived exosomes may offer significant advantages over traditional MSC-based therapies in regenerative medicine. By using exosomes, practitioners can avoid potential risks associated with MSC-centered therapies such as allogeneic immunological rejection, unwanted differentiation, and obstruction of small vessels caused by intravenous MSC injection. These benefits are critical for optimizing treatment outcomes [4].

  • ophthalmology
  • ocular pharmacology
  • anterior segment diseases
  • posterior segment diseases
  • cell-based drug delivery systems
  • MSCs-based cell therapy
  • MSC-derived exosome
  • exosomes-baseddrug delivery
  • tissue repair and regeneration

Overcoming Challenges in the Clinical Translation of MSC-Exosomes

Despite the potential of MSC-exosome therapies, unresolved challenges such as non-uniformity in isolation and purification, unclear mechanisms of action, low-yield capacity, and unstandardized large-scale production protocols still need to be addressed [123]. However, the most significant obstacle to clinical transformation is the lack of standardized quality assessment criteria and the heterogeneity of exosomal products. The International Society for Cell and Gene Therapy has developed criteria differentiating MSCs sourced from various targets [124], but more comprehensive criteria are needed to address parental MSC quality, MSC-exosome quality, and potential for ex vivo expansion. Whereas, heterogeneity hinders product quality and management, reducing reproducibility in both in vivo and in vitro contexts.

 

Assessment of Parental MSCs as Proxy Indicator of MSC-Exosome Quality

To ensure high-quality MSC-exosome products, a standardized donor selection and screening approach is needed. Studies have investigated various assessment metrics, such as in vitro characteristics, donor demographics, EV sourcing, and genetic biomarkers, to differentiate high- and low-quality MSCs. These criteria can be used as proxy indicators of MSC-exosome quality and inform clinical decision-making. High-potency MSCs are believed to be more effective, but a comprehensive assessment panel is required for informed decision-making.

Samsonraj et al. (2015) investigated factors affecting bone marrow-derived mesenchymal stem cells (BMSCs) and their in vivo tissue regeneration potential [130]. MSCs with high colony-forming unit-fibroblasts (CFU-F) showed an increase in small-sized cells with lengthened telomeres, and high growth capacity performed better on ectopic bone-formation assays. STRO-1+ and nestin+ MSCs expressing PDGFR-α were predictive of greater high-growth and colony formation capacity. Additionally, a global gene expression analysis revealed that low-growth BMSCs had more maturation-based cellular processes, while high-growth BMSCs had more proliferation-based processes.

Sathiyanathan et al. (2020) conducted a transcriptomic analysis of genetic biomarkers in BMSCs to understand their effect on scalability [131]. The study identified glutathione S-transferase theta 1 (GSTT1) as the most differentially expressed gene, with low-growth BMSCs showing a fifty-fold greater expression of the gene. High-growth BMSCs were found to have a genomic deletion of GSTT1, leading to its repression. The subsequent double-blind study revealed that GSTT1-null BMSCs demonstrated greater growth and self-renewal capacity, longer telomeres, and higher total cell count and CFU-F efficiency, making GSTT1 a meaningful genetic biomarker for BMSC scalability. Therefore, GSTT1 status in donors could be used as a rapid assessment tool for harvesting-related decisions and to inform usage in clinical applications.

Boulestreau et al. (2020) reviewed the impact of aging on MSC quality [132]. Age-related changes to MSCs result in the loss or increased dysfunctionality of stem cell functionality. While the precise etiology of the relationship between MSC functionality and age remains unclear, some notable findings have been consistently reported in the literature. First, the proliferative and clonogenic capacity of BMSCs is negatively associated with age [133], with specific cell-surface markers such as decreased CD146 expression and upregulated CD296 linked to late-passage MSCs. Second, reactive oxygen species and consequent oxidative stress are higher in aging MSCs. Some studies have investigated possible treatments for reversing age-related changes to MSCs. For example, melatonin is protective against oxidative stress and senescence [138], and selective inhibitor ML141 decreased CDC42 protein activity in aging MSCs [139]. Siegal et al. (2013) found that younger female donors exhibited higher clonogenicity and increased proliferative rates and had more favorable BMSCs than other donor demographics [140].

Ulum et al. (2018) reported that high BMI associated with obesity predicts reduced MSC quality [141]. In donors with higher BMI, BMSCs exhibited functional impairment, including a significant reduction in osteogenic differentiation and slower proliferation rates. These changes were accompanied by a higher proportion of senescent cells and a decline in critical stromal adhesion proteins and MSC markers. The proposed mechanism for this reduction in quality is the promotion of ER stress-related genes (ATF4 and CHOP) due to protein misfolding in obese individuals. This leads to the unfolded protein response (UPR) and subsequently, stem cell dysfunction. To address these challenges, potential solutions have been investigated, including treatment with TUDCA and 4-PBA, both of which regulate UPR- and ER stress-related proteins, improve osteogenic and adipogenic differentiation, and prevent UPR dysfunction. These treatments may attenuate the obesity-related decrease in MSC quality, as the global obesity rate continues to rise.

Li et al. (2021) compared the benefits and drawbacks of allogeneic and autologous MSCs as extracellular vesicle sources [142]. Although the literature is still debating the best source, allogeneic MSCs are increasingly used in MSC-based therapies due to their favorable safety profile, high accessibility, and donor selection. However, potential immune rejection and donor heterogeneity may limit its application. Autologous MSC sourcing is safer as it is sourced directly from the patient, but the primary disadvantages are long-time availability and potential disease-candidate genes. Additionally, the local microenvironment can influence the functional properties of MSCs; thus, tissue-derived MSCs from a potentially harmful microenvironment may not be ideal. To optimize treatment outcomes and patient satisfaction, clinicians should thoroughly communicate the safety and efficacy profile of each approach to their patients.

 

Overcoming the Barriers of MSC-Exosome Heterogeneity

Parental and exosomal heterogeneity hinder the quality and management of MSC-exosome products, reducing their reproducibility in in vivo and in vitro contexts [125], leading to heterogeneous results. Different parental sources have shown varying therapeutic effects. For example, BMSC-derived MSC-exosomes are four times superior to ADSC-derived exosomes in terms of angiogenicity, while endometrial-derived MSC-exosomes are significantly better than both. ADSC-derived exosomes have been shown to produce more cardio-protective factors such as VEGF and HGF. Regarding immunomodulation, BMSC- and ADSC-derived exosomes can induce M2 polarization of macrophages, but BMSCs show a 3.2-fold increase in CD206 expression compared to only 1.5-fold in ADSCs. Given such heterogeneity, further research is needed to explore the nuanced effects of different parental sources and corresponding exosomes to determine the optimal sourcing and extraction protocol for patients. Additionally, comprehensive criteria for assessing the quality of parental MSCs, MSC-exosomes, and their potential for ex vivo expansion are needed for exosome-based therapies to be clinically successful [123, 124].

Kou et al. (2020) suggest a solution to the problem of source and exosome product heterogeneity: extracting exosome products from human pluripotent stem cells (hPSCs)-derived MSCs [125]. Clinical trials exploring the use of hPSC-derived MSC-exosomes in patients with refractory graft-versus-host disease (GVHD) have shown promising results, with significant improvements in cutaneous chronic GVHD and rejection following abdominal organ transplantation. The hPSC-MSCs have a higher passage number and can produce higher exosomal yields than traditional MSCs. Additionally, hPSC-MSCs have improved secretion and amplification abilities, resulting in higher quality MSC-exosome products and cost-efficient, large-scale production potential.

Varderidou-Minasian and Lorenowicz (2020) highlight that the paracrine signaling of MSC therapy is crucial, rather than engraftment or differentiation capabilities of the transplanted MSCs [126]. Therefore, the qualitative and quantitative characteristics of MSC-secretomes, including EVs and exosomes, should be considered during quality assessment. Recent advancements in cell culture technology indicate that 3D culture systems better replicate in vivo conditions than traditional 2D cultures, preserving critical traits such as morphology, functionality, and structure, which are important for proliferative and differentiation capacity, ultimately enhancing exosomal efficacy [127, 128].

Ni Su et al. (2017) found that extracellular matrices with oriented fibers, compared to non-oriented, were more conducive to the release of anti-inflammatory and angiogenic-promoting factors [129]. Therefore, 3D culturing is a better option for replicating in vivo conditions than traditional static adherent 2D cultures, which hamper exosomal efficacy [127]. 3D culturing can be divided into material-free and material-supported, with the latter more conducive for cell-to-cell connectivity and signaling. Examples of material-free cell cultures include hydrogel-assisted 3D cultures and scaffold-free suspension cultures, while material-supported cultures include hollow fiber bioreactors. The latter have demonstrated a 19.4-greater yield than 2D cultures within shorter culture periods. MSC-exosomes harvested from 3D cell cultures have also been shown to improve several conditions in rat models, including improved angiogenicity and the proliferation and migration of endothelial cells in the context of injury repair.

Preconditioning of MSCs is another approach to improve the quality of MSC-exosome products [127]. Hypoxic preconditioning, involving low-oxygen tension, enhances the MSCs' proliferative capacity and genetic stability, leading to improved migratory and paracrine capacity. The extracted exosomes from these preconditioned MSCs, called Hyp-MSC-Exos, have shown therapeutic benefits in various diseases, such as CNS issues and diabetic wound healing. Another preconditioning protocol, cytokine preconditioning, involves the stimulation of cytokines and inflammatory factors, which increases the paracrine efficiency of MSC-exosomes. Exposure to TNF-α, IL-1β, or IFN-γ promotes the release of exosomes with increased anti-inflammatory properties and the presence of inflammation-suppressing miRNAs. Additionally, chemical and physical preconditioning approaches, such as treatment with metformin or monochromatic blue light (451 nm), can also produce similar effects.

 

[1][2][3][4][5][6][7][8][9][10][11][12][13][14][15][16][17][18][19][20][21][22][23][24][25][26][27][28][29][30][31][32][33][34][35][36][37][38][39][40][41][42][43][44][45][46][47][48][49][50][51][52][53][54][55][56][57][58][59][60][61][62][63][64][65][66][67][68][69][70][71][72][73][74][75][76][77][78][79][80][81][82][83][84][85][86][87][88][89][90][91][92][93][94][95][96][97][98][99][100][101][102][103][104][105][106][107][108][109][110][111][112][113][114][115][116][117][118][119][120][121][122][123][124][125][126][127][128][129][130][131][132][133][134][135][136][137][138][139][140][141][142]

This entry is adapted from the peer-reviewed paper 10.3390/pharmaceutics15041167

References

  1. Niamprem, P.; Srinivas, S.P.; Tiyaboonchai, W. Penetration of Nile Red-Loaded Nanostructured Lipid Carriers (NLCs) across the Porcine Cornea. Colloids Surf. B Biointerfaces 2019, 176, 371–378. [Google Scholar] [CrossRef] [PubMed]
  2. Blass, S.; Teubl, B.; Fröhlich, E.; Meindl, C.; Rabensteiner, D.F.; Trummer, G.; Schmut, O.; Zimmer, A.; Roblegg, E. Permeability Studies on the Ocular Absorbance of Nanostructured Materials Across the Cornea. Sci. Pharm. 2010, 78, 678. [Google Scholar] [CrossRef][Green Version]
  3. Mohammadpour, M.; Hashemi, H.; Jabbarvand, M.; Delrish, E. Penetration of Silicate Nanoparticles into the Corneal Stroma and Intraocular Fluids. Cornea 2014, 33, 738. [Google Scholar] [CrossRef]
  4. Yu, B.; Shao, H.; Su, C.; Jiang, Y.; Chen, X.; Bai, L.; Zhang, Y.; Li, Q.; Zhang, X.; Li, X. Exosomes Derived from MSCs Ameliorate Retinal Laser Injury Partially by Inhibition of MCP-1. Sci. Rep. 2016, 6, 34562. [Google Scholar] [CrossRef] [PubMed][Green Version]
  5. Yu, B.; Li, X.-R.; Zhang, X.-M. Mesenchymal Stem Cell-Derived Extracellular Vesicles as a New Therapeutic Strategy for Ocular Diseases. World J. Stem Cells 2020, 12, 178–187. [Google Scholar] [CrossRef] [PubMed]
  6. Cui, Y.; Liu, C.; Huang, L.; Chen, J.; Xu, N. Protective Effects of Intravitreal Administration of Mesenchymal Stem Cell-Derived Exosomes in an Experimental Model of Optic Nerve Injury. Exp. Cell Res. 2021, 407, 112792. [Google Scholar] [CrossRef] [PubMed]
  7. Zhang, W.; Wang, Y.; Kong, Y. Exosomes Derived from Mesenchymal Stem Cells Modulate MiR-126 to Ameliorate Hyperglycemia-Induced Retinal Inflammation Via Targeting HMGB1. Investig. Opthalmol. Vis. Sci. 2019, 60, 294. [Google Scholar] [CrossRef][Green Version]
  8. Xu, H.-K.; Chen, L.-J.; Zhou, S.-N.; Li, Y.-F.; Xiang, C. Multifunctional Role of MicroRNAs in Mesenchymal Stem Cell-Derived Exosomes in Treatment of Diseases. World J. Stem Cells 2020, 12, 1276–1294. [Google Scholar] [CrossRef]
  9. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Théry, C. Specificities of Secretion and Uptake of Exosomes and Other Extracellular Vesicles for Cell-to-Cell Communication. Nat. Cell Biol. 2019, 21, 9–17. [Google Scholar] [CrossRef]
  10. Van Niel, G.; D’Angelo, G.; Raposo, G. Shedding Light on the Cell Biology of Extracellular Vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
  11. Xu, M.; Ji, J.; Jin, D.; Wu, Y.; Wu, T.; Lin, R.; Zhu, S.; Jiang, F.; Ji, Y.; Bao, B.; et al. The Biogenesis and Secretion of Exosomes and Multivesicular Bodies (MVBs): Intercellular Shuttles and Implications in Human Diseases. Genes Dis. 2022, S2352304222000976. [Google Scholar] [CrossRef]
  12. Wu, H.; Turner, C.; Gardner, J.; Temple, B.; Brennwald, P. The Exo70 Subunit of the Exocyst Is an Effector for Both Cdc42 and Rho3 Function in Polarized Exocytosis. Mol. Biol. Cell 2010, 21, 430–442. [Google Scholar] [CrossRef] [PubMed][Green Version]
  13. Hung, M.E.; Leonard, J.N. Stabilization of Exosome-Targeting Peptides via Engineered Glycosylation. J. Biol. Chem. 2015, 290, 8166–8172. [Google Scholar] [CrossRef][Green Version]
  14. McKelvey, K.J.; Powell, K.L.; Ashton, A.W.; Morris, J.M.; McCracken, S.A. Exosomes: Mechanisms of Uptake. J. Circ. Biomark. 2015, 4, 7. [Google Scholar] [CrossRef][Green Version]
  15. Bian, B.; Zhao, C.; He, X.; Gong, Y.; Ren, C.; Ge, L.; Zeng, Y.; Li, Q.; Chen, M.; Weng, C.; et al. Exosomes Derived from Neural Progenitor Cells Preserve Photoreceptors during Retinal Degeneration by Inactivating Microglia. J. Extracell. Vesicles 2020, 9, 1748931. [Google Scholar] [CrossRef] [PubMed][Green Version]
  16. Yeo, R.W.Y.; Lai, R.C.; Zhang, B.; Tan, S.S.; Yin, Y.; Teh, B.J.; Lim, S.K. Mesenchymal Stem Cell: An Efficient Mass Producer of Exosomes for Drug Delivery. Adv. Drug Deliv. Rev. 2013, 65, 336–341. [Google Scholar] [CrossRef]
  17. Liu, X.; Hu, L.; Liu, F. Mesenchymal Stem Cell-Derived Extracellular Vesicles for Cell-Free Therapy of Ocular Diseases. Extracell. Vesicles Circ. Nucleic Acids 2022, 3, 102–117. [Google Scholar] [CrossRef]
  18. Samaeekia, R.; Rabiee, B.; Putra, I.; Shen, X.; Park, Y.J.; Hematti, P.; Eslani, M.; Djalilian, A.R. Effect of Human Corneal Mesenchymal Stromal Cell-Derived Exosomes on Corneal Epithelial Wound Healing. Investig. Opthalmol. Vis. Sci. 2018, 59, 5194. [Google Scholar] [CrossRef][Green Version]
  19. Zhang, Z.; Mugisha, A.; Fransisca, S.; Liu, Q.; Xie, P.; Hu, Z. Emerging Role of Exosomes in Retinal Diseases. Front. Cell Dev. Biol. 2021, 9, 643680. [Google Scholar] [CrossRef]
  20. Boukouris, S.; Mathivanan, S. Exosomes in Bodily Fluids Are a Highly Stable Resource of Disease Biomarkers. PROTEOMICS Clin. Appl. 2015, 9, 358–367. [Google Scholar] [CrossRef][Green Version]
  21. Chen, T.S.; Lai, R.C.; Lee, M.M.; Choo, A.B.H.; Lee, C.N.; Lim, S.K. Mesenchymal Stem Cell Secretes Microparticles Enriched in Pre-MicroRNAs. Nucleic Acids Res. 2010, 38, 215–224. [Google Scholar] [CrossRef] [PubMed][Green Version]
  22. Lai, R.C.; Tan, S.S.; Teh, B.J.; Sze, S.K.; Arslan, F.; de Kleijn, D.P.; Choo, A.; Lim, S.K. Proteolytic Potential of the MSC Exosome Proteome: Implications for an Exosome-Mediated Delivery of Therapeutic Proteasome. Int. J. Proteomics 2012, 2012, 971907. [Google Scholar] [CrossRef] [PubMed][Green Version]
  23. Glover, K.; Mishra, D.; Singh, T.R.R. Epidemiology of Ocular Manifestations in Autoimmune Disease. Front. Immunol. 2021, 12, 744396. [Google Scholar] [CrossRef] [PubMed]
  24. Seo, Y.; Kim, H.-S.; Hong, I.-S. Stem Cell-Derived Extracellular Vesicles as Immunomodulatory Therapeutics. Available online: https://www.hindawi.com/journals/sci/2019/5126156/ (accessed on 14 February 2023).
  25. Kuriyan, A.E.; Albini, T.A.; Townsend, J.H.; Rodriguez, M.; Pandya, H.K.; Leonard, R.E.; Parrott, M.B.; Rosenfeld, P.J.; Flynn, H.W.; Goldberg, J.L. Vision Loss after Intravitreal Injection of Autologous “Stem Cells” for AMD. N. Engl. J. Med. 2017, 376, 1047–1053. [Google Scholar] [CrossRef][Green Version]
  26. Sun, H.; Pratt, R.E.; Hodgkinson, C.P.; Dzau, V.J. Sequential Paracrine Mechanisms Are Necessary for the Therapeutic Benefits of Stem Cell Therapy. Am. J. Physiol. Cell Physiol. 2020, 319, C1141–C1150. [Google Scholar] [CrossRef]
  27. Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, Biologic Function and Clinical Potential. Cell Biosci. 2019, 9, 19. [Google Scholar] [CrossRef]
  28. Liang, Y.; Duan, L.; Lu, J.; Xia, J. Engineering Exosomes for Targeted Drug Delivery. Theranostics 2021, 11, 3183–3195. [Google Scholar] [CrossRef]
  29. Seyedrazizadeh, S.-Z.; Poosti, S.; Nazari, A.; Alikhani, M.; Shekari, F.; Pakdel, F.; Shahpasand, K.; Satarian, L.; Baharvand, H. Extracellular Vesicles Derived from Human ES-MSCs Protect Retinal Ganglion Cells and Preserve Retinal Function in a Rodent Model of Optic Nerve Injury. Stem Cell Res. Ther. 2020, 11, 203. [Google Scholar] [CrossRef]
  30. Pan, D.; Chang, X.; Xu, M.; Zhang, M.; Zhang, S.; Wang, Y.; Luo, X.; Xu, J.; Yang, X.; Sun, X. UMSC-Derived Exosomes Promote Retinal Ganglion Cells Survival in a Rat Model of Optic Nerve Crush. J. Chem. Neuroanat. 2019, 96, 134–139. [Google Scholar] [CrossRef]
  31. Chen, C.C.; Liu, L.; Ma, F.; Wong, C.W.; Guo, X.E.; Chacko, J.V.; Farhoodi, H.P.; Zhang, S.X.; Zimak, J.; Ségaliny, A.; et al. Elucidation of Exosome Migration across the Blood-Brain Barrier Model In Vitro. Cell. Mol. Bioeng. 2016, 9, 509–529. [Google Scholar] [CrossRef][Green Version]
  32. Li, C.; Qin, S.; Wen, Y.; Zhao, W.; Huang, Y.; Liu, J. Overcoming the Blood-Brain Barrier: Exosomes as Theranostic Nanocarriers for Precision Neuroimaging. J. Control. Release Off. J. Control. Release Soc. 2022, 349, 902–916. [Google Scholar] [CrossRef] [PubMed]
  33. Heidarzadeh, M.; Gürsoy-Özdemir, Y.; Kaya, M.; Eslami Abriz, A.; Zarebkohan, A.; Rahbarghazi, R.; Sokullu, E. Exosomal Delivery of Therapeutic Modulators through the Blood–Brain Barrier; Promise and Pitfalls. Cell Biosci. 2021, 11, 142. [Google Scholar] [CrossRef] [PubMed]
  34. Elliott, R.O.; He, M. Unlocking the Power of Exosomes for Crossing Biological Barriers in Drug Delivery. Pharmaceutics 2021, 13, 122. [Google Scholar] [CrossRef] [PubMed]
  35. Tang, Y.; Zhou, Y.; Li, H.-J. Advances in Mesenchymal Stem Cell Exosomes: A Review. Stem Cell Res. Ther. 2021, 12, 71. [Google Scholar] [CrossRef] [PubMed]
  36. Jia, Y.; Ni, Z.; Sun, H.; Wang, C. Microfluidic Approaches Toward the Isolation and Detection of Exosome Nanovesicles. IEEE Access 2019, 7, 45080–45098. [Google Scholar] [CrossRef]
  37. Zhang, Y.; Bi, J.; Huang, J.; Tang, Y.; Du, S.; Li, P. Exosome: A Review of Its Classification, Isolation Techniques, Storage, Diagnostic and Targeted Therapy Applications. Int. J. Nanomed. 2020, 15, 6917–6934. [Google Scholar] [CrossRef]
  38. Moisseiev, E.; Anderson, J.D.; Oltjen, S.; Goswami, M.; Zawadzki, R.J.; Nolta, J.A.; Park, S.S. Protective Effect of Intravitreal Administration of Exosomes Derived from Mesenchymal Stem Cells on Retinal Ischemia. Curr. Eye Res. 2017, 42, 1358–1367. [Google Scholar] [CrossRef][Green Version]
  39. Zhou, T.; He, C.; Lai, P.; Yang, Z.; Liu, Y.; Xu, H.; Lin, X.; Ni, B.; Ju, R.; Yi, W.; et al. MiR-204–Containing Exosomes Ameliorate GVHD-Associated Dry Eye Disease. Sci. Adv. 2022, 8, eabj9617. [Google Scholar] [CrossRef]
  40. Wang, J.; Chen, D.; Ho, E.A. Challenges in the Development and Establishment of Exosome-Based Drug Delivery Systems. J. Control. Release 2021, 329, 894–906. [Google Scholar] [CrossRef]
  41. Sun, Y.; Liu, G.; Zhang, K.; Cao, Q.; Liu, T.; Li, J. Mesenchymal Stem Cells-Derived Exosomes for Drug Delivery. Stem Cell Res. Ther. 2021, 12, 561. [Google Scholar] [CrossRef]
  42. Yu, M.; Liu, W.; Li, J.; Lu, J.; Lu, H.; Jia, W.; Liu, F. Exosomes Derived from Atorvastatin-Pretreated MSC Accelerate Diabetic Wound Repair by Enhancing Angiogenesis via AKT/ENOS Pathway. Stem Cell Res. Ther. 2020, 11, 350. [Google Scholar] [CrossRef]
  43. Wilson, S.E. Corneal Wound Healing. Exp. Eye Res. 2020, 197, 108089. [Google Scholar] [CrossRef]
  44. Du, Y.; SundarRaj, N.; Funderburgh, M.L.; Harvey, S.A.; Birk, D.E.; Funderburgh, J.L. Secretion and Organization of a Cornea-like Tissue In Vitro by Stem Cells from Human Corneal Stroma. Investig. Ophthalmol. Vis. Sci. 2007, 48, 5038–5045. [Google Scholar] [CrossRef] [PubMed]
  45. Sharif, Z.; Sharif, W. Corneal Neovascularization: Updates on Pathophysiology, Investigations & Management. Rom. J. Ophthalmol. 2019, 63, 15–22. [Google Scholar] [PubMed]
  46. Yu, B.; Zhang, X.; Li, X. Exosomes Derived from Mesenchymal Stem Cells. Int. J. Mol. Sci. 2014, 15, 4142–4157. [Google Scholar] [CrossRef] [PubMed][Green Version]
  47. Phinney, D.G.; Pittenger, M.F. Concise Review: MSC-Derived Exosomes for Cell-Free Therapy. Stem Cells 2017, 35, 851–858. [Google Scholar] [CrossRef][Green Version]
  48. Tao, H.; Chen, X.; Cao, H.; Zheng, L.; Li, Q.; Zhang, K.; Han, Z.; Han, Z.-C.; Guo, Z.; Li, Z.; et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles for Corneal Wound Repair. Stem Cells Int. 2019, 2019, 5738510. [Google Scholar] [CrossRef][Green Version]
  49. Yu, Z.; Hao, R.; Du, J.; Wu, X.; Chen, X.; Zhang, Y.; Li, W.; Gu, Z.; Yang, H. A Human Cornea-on-a-Chip for the Study of Epithelial Wound Healing by Extracellular Vesicles. iScience 2022, 25, 104200. [Google Scholar] [CrossRef]
  50. Liu, X.; Li, X.; Wu, G.; Qi, P.; Zhang, Y.; Liu, Z.; Li, X.; Yu, Y.; Ye, X.; Li, Y.; et al. Umbilical Cord Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Deliver MiR-21 to Promote Corneal Epithelial Wound Healing through PTEN/PI3K/Akt Pathway. Stem Cells Int. 2022, 2022, 1252557. [Google Scholar] [CrossRef]
  51. Ma, S.; Yin, J.; Hao, L.; Liu, X.; Shi, Q.; Diao, Y.; Yu, G.; Liu, L.; Chen, J.; Zhong, J. Exosomes from Human Umbilical Cord Mesenchymal Stem Cells Treat Corneal Injury via Autophagy Activation. Front. Bioeng. Biotechnol. 2022, 10, 879192. [Google Scholar] [CrossRef]
  52. Shen, T.; Zheng, Q.-Q.; Shen, J.; Li, Q.-S.; Song, X.-H.; Luo, H.-B.; Hong, C.-Y.; Yao, K. Effects of Adipose-Derived Mesenchymal Stem Cell Exosomes on Corneal Stromal Fibroblast Viability and Extracellular Matrix Synthesis. Chin. Med. J. 2018. Available online: https://mednexus.org/doi/full/10.4103/0366-6999.226889 (accessed on 12 February 2023). [CrossRef] [PubMed]
  53. Du, Y.; Funderburgh, M.L.; Mann, M.M.; SundarRaj, N.; Funderburgh, J.L. Multipotent Stem Cells in Human Corneal Stroma. Stem Cells 2005, 23, 1266–1275. Available online: https://academic.oup.com/stmcls/article/23/9/1266/6399870 (accessed on 12 February 2023). [CrossRef] [PubMed][Green Version]
  54. Du, Y.; Carlson, E.C.; Funderburgh, M.L.; Birk, D.E.; Pearlman, E.; Guo, N.; Kao, W.W.-Y.; Funderburgh, J.L. Stem Cell Therapy Restores Transparency to Defective Murine Corneas. Stem Cells 2009, 27, 1635–1642. Available online: https://academic.oup.com/stmcls/article/27/7/1635/6402401 (accessed on 12 February 2023). [CrossRef][Green Version]
  55. Wang, Y.; Gao, G.; Wu, Y.; Wang, Y.; Wu, X.; Zhou, Q. S100A4 Silencing Facilitates Corneal Wound Healing After Alkali Burns by Promoting Autophagy via Blocking the PI3K/Akt/MTOR Signaling Pathway. Investig. Ophthalmol. Vis. Sci. 2020, 61, 19. [Google Scholar] [CrossRef] [PubMed]
  56. Li, Y.; Jin, R.; Li, L.; Choi, J.S.; Kim, J.; Yoon, H.J.; Park, J.H.; Yoon, K.C. Blue Light Induces Impaired Autophagy through Nucleotide-Binding Oligomerization Domain 2 Activation on the Mouse Ocular Surface. Int. J. Mol. Sci. 2021, 22, 2015. [Google Scholar] [CrossRef] [PubMed]
  57. Tang, Q.; Lu, B.; He, J.; Chen, X.; Fu, Q.; Han, H.; Luo, C.; Yin, H.; Qin, Z.; Lyu, D.; et al. Exosomes-Loaded Thermosensitive Hydrogels for Corneal Epithelium and Stroma Regeneration. Biomaterials 2022, 280, 121320. [Google Scholar] [CrossRef] [PubMed]
  58. Sun, X.; Song, W.; Teng, L.; Huang, Y.; Liu, J.; Peng, Y.; Lu, X.; Yuan, J.; Zhao, X.; Zhao, Q.; et al. MiRNA 24-3p-Rich Exosomes Functionalized DEGMA-Modified Hyaluronic Acid Hydrogels for Corneal Epithelial Healing. Biocative Mater. 2023, 25, 640–656. Available online: https://www.sciencedirect.com/science/article/pii/S2452199X22003097?via%3Dihub (accessed on 12 February 2023). [CrossRef]
  59. Lin, H.; Yiu, S.C. Dry Eye Disease: A Review of Diagnostic Approaches and Treatments. Saudi J. Ophthalmol. Off. J. Saudi Ophthalmol. Soc. 2014, 28, 173–181. [Google Scholar] [CrossRef][Green Version]
  60. Wu, K.Y.; Chen, W.T.; Chu-Bédard, Y.-K.; Patel, G.; Tran, S.D. Management of Sjogren’s Dry Eye Disease—Advances in Ocular Drug Delivery Offering a New Hope. Pharmaceutics 2023, 15, 147. [Google Scholar] [CrossRef]
  61. Lai, P.; Chen, X.; Guo, L.; Wang, Y.; Liu, X.; Liu, Y.; Zhou, T.; Huang, T.; Geng, S.; Luo, C.; et al. A Potent Immunomodulatory Role of Exosomes Derived from Mesenchymal Stromal Cells in Preventing CGVHD. J. Hematol. Oncol. 2018, 11, 135. [Google Scholar] [CrossRef][Green Version]
  62. Zhang, B.; Yeo, R.W.Y.; Lai, R.C.; Sim, E.W.K.; Chin, K.C.; Lim, S.K. Mesenchymal Stromal Cell Exosome–Enhanced Regulatory T-Cell Production through an Antigen-Presenting Cell–Mediated Pathway. Cytotherapy 2018, 20, 687–696. [Google Scholar] [CrossRef] [PubMed]
  63. Guo, R.; Liang, Q.; He, Y.; Wang, C.; Jiang, J.; Chen, T.; Zhang, D.; Hu, K. Mesenchymal Stromal Cells-Derived Extracellular Vesicles Regulate Dendritic Cell Functions in Dry Eye Disease. Cells 2023, 12, 33. [Google Scholar] [CrossRef] [PubMed]
  64. Wang, G.; Li, H.; Long, H.; Gong, X.; Hu, S.; Gong, C. Exosomes Derived from Mouse Adipose-Derived Mesenchymal Stem Cells Alleviate Benzalkonium Chloride-Induced Mouse Dry Eye Model via Inhibiting NLRP3 Inflammasome. Ophthalmic Res. 2022, 65, 40–51. [Google Scholar] [CrossRef] [PubMed]
  65. Yu, C.; Chen, P.; Xu, J.; Liu, Y.; Li, H.; Wang, L.; Di, G. HADSCs Derived Extracellular Vesicles Inhibit NLRP3 inflammasome Activation and Dry Eye. Sci. Rep. 2020, 10, 14521. [Google Scholar] [CrossRef]
  66. Ma, F.; Feng, J.; Liu, X.; Tian, Y.; Wang, W.-J.; Luan, F.-X.; Wang, Y.-J.; Yang, W.-Q.; Bai, J.-Y.; Zhang, Y.-Q.; et al. Ascorbic Acid-Coupled Mesenchymal Stem Cell-Derived Exosomes Ameliorate Dry Eye Disease. Preprints 2020, 2020060316. [Google Scholar] [CrossRef]
  67. Study Record|Beta ClinicalTrials.Gov. Available online: https://beta.clinicaltrials.gov/study/NCT04213248?tab=results (accessed on 13 February 2023).
  68. Zhao, J.; An, Q.; Zhu, X.; Yang, B.; Gao, X.; Niu, Y.; Zhang, L.; Xu, K.; Ma, D. Research Status and Future Prospects of Extracellular Vesicles in Primary Sjögren’s Syndrome. Stem Cell Res. Ther. 2022, 13, 230. [Google Scholar] [CrossRef]
  69. Gong, B.; Zheng, L.; Lu, Z.; Huang, J.; Pu, J.; Pan, S.; Zhang, M.; Liu, J.; Tang, J. Mesenchymal Stem Cells Negatively Regulate CD4+ T Cell Activation in Patients with Primary Sjögren Syndrome through the MiRNA-125b and MiRNA-155 TCR Pathway. Mol. Med. Rep. 2020, 23, 43. [Google Scholar] [CrossRef]
  70. Li, B.; Xing, Y.; Gan, Y.; He, J.; Hua, H. Labial Gland-Derived Mesenchymal Stem Cells and Their Exosomes Ameliorate Murine Sjögren’s Syndrome by Modulating the Balance of Treg and Th17 Cells. Stem Cell Res. Ther. 2021, 12, 478. Available online: https://stemcellres.biomedcentral.com/articles/10.1186/s13287-021-02541-0 (accessed on 13 February 2023). [CrossRef]
  71. Lind, E.F.; Ohashi, P.S. Mir-155, a Central Modulator of T-Cell Responses: Highlights. Eur. J. Immunol. 2014, 44, 11–15. [Google Scholar] [CrossRef]
  72. Rui, K.; Hong, Y.; Zhu, Q.; Shi, X.; Xiao, F.; Fu, H.; Yin, Q.; Xing, Y.; Wu, X.; Kong, X.; et al. Olfactory Ecto-Mesenchymal Stem Cell-Derived Exosomes Ameliorate Murine Sjögren’s Syndrome by Modulating the Function of Myeloid-Derived Suppressor Cells. Cell. Mol. Immunol. 2021, 18, 440–451. [Google Scholar] [CrossRef]
  73. Tomatsu, S.; Pitz, S.; Hampel, U. Ophthalmological Findings in Mucopolysaccharidoses. J. Clin. Med. 2019, 8, 1467. [Google Scholar] [CrossRef] [PubMed][Green Version]
  74. Coulson-Thomas, V.J.; Caterson, B.; Kao, W.W.-Y. Transplantation of Human Umbilical Mesenchymal Stem Cells Cures the Corneal Defects of Mucopolysaccharidosis VII Mice. Stem Cells 2013, 31, 2116–2126. Available online: https://academic.oup.com/stmcls/article/31/10/2116/6408126 (accessed on 13 February 2023). [CrossRef] [PubMed][Green Version]
  75. Flanagan, M.; Pathak, I.; Gan, Q.; Winter, L.; Emnet, R.; Akel, S.; Montaño, A.M. Umbilical Mesenchymal Stem Cell-Derived Extracellular Vesicles as Enzyme Delivery Vehicle to Treat Morquio a Fibroblasts. Stem Cell Res. Ther. 2021, 12, 276. [Google Scholar] [CrossRef] [PubMed]
  76. Doozandeh, A.; Yazdani, S. Neuroprotection in Glaucoma. J. Ophthalmic Vis. Res. 2016, 11, 209–220. [Google Scholar] [CrossRef] [PubMed][Green Version]
  77. Mead, B.; Tomarev, S. Bone Marrow-Derived Mesenchymal Stem Cells-Derived Exosomes Promote Survival of Retinal Ganglion Cells Through MiRNA-Dependent Mechanisms. Stem Cells Transl. Med. 2017, 6, 1273–1285. [Google Scholar] [CrossRef] [PubMed]
  78. Mead, B.; Ahmed, Z.; Tomarev, S. Mesenchymal Stem Cell–Derived Small Extracellular Vesicles Promote Neuroprotection in a Genetic DBA/2J Mouse Model of Glaucoma. Investig. Opthalmol. Vis. Sci. 2018, 59, 5473. [Google Scholar] [CrossRef] [PubMed][Green Version]
  79. Mead, B.; Amaral, J.; Tomarev, S. Mesenchymal Stem Cell–Derived Small Extracellular Vesicles Promote Neuroprotection in Rodent Models of Glaucoma. Investig. Opthalmol. Vis. Sci. 2018, 59, 702. [Google Scholar] [CrossRef]
  80. Mead, B.; Chamling, X.; Zack, D.J.; Ahmed, Z.; Tomarev, S. TNFα-Mediated Priming of Mesenchymal Stem Cells Enhances Their Neuroprotective Effect on Retinal Ganglion Cells. Investig. Opthalmol. Vis. Sci. 2020, 61, 6. [Google Scholar] [CrossRef][Green Version]
  81. Park, M.; Shin, H.A.; Duong, V.-A.; Lee, H.; Lew, H. The Role of Extracellular Vesicles in Optic Nerve Injury: Neuroprotection and Mitochondrial Homeostasis. Cells 2022, 11, 3720. [Google Scholar] [CrossRef]
  82. Berry, M.; Ahmed, Z.; Morgan-Warren, P.; Fulton, D.; Logan, A. Prospects for MTOR-Mediated Functional Repair after Central Nervous System Trauma. Neurobiol. Dis. 2016, 85, 99–110. [Google Scholar] [CrossRef][Green Version]
  83. Park, K.K.; Liu, K.; Hu, Y.; Smith, P.D.; Wang, C.; Cai, B.; Xu, B.; Connolly, L.; Kramvis, I.; Sahin, M.; et al. Promoting Axon Regeneration in the Adult CNS by Modulation of the PTEN/MTOR Pathway. Science 2008, 322, 963–966. [Google Scholar] [CrossRef] [PubMed][Green Version]
  84. Katakowski, M.; Buller, B.; Zheng, X.; Lu, Y.; Rogers, T.; Osobamiro, O.; Shu, W.; Jiang, F.; Chopp, M. Exosomes from Marrow Stromal Cells Expressing MiR-146b Inhibit Glioma Growth. Cancer Lett. 2013, 335, 201–204. [Google Scholar] [CrossRef] [PubMed][Green Version]
  85. Douglas, M.R.; Morrison, K.C.; Jacques, S.J.; Leadbeater, W.E.; Gonzalez, A.M.; Berry, M.; Logan, A.; Ahmed, Z. Off-Target Effects of Epidermal Growth Factor Receptor Antagonists Mediate Retinal Ganglion Cell Disinhibited Axon Growth. Brain 2009, 132, 3102–3121. [Google Scholar] [CrossRef][Green Version]
  86. Koprivica, V.; Cho, K.-S.; Park, J.B.; Yiu, G.; Atwal, J.; Gore, B.; Kim, J.A.; Lin, E.; Tessier-Lavigne, M.; Chen, D.F.; et al. EGFR Activation Mediates Inhibition of Axon Regeneration by Myelin and Chondroitin Sulfate Proteoglycans. Science 2005, 310, 106–110. [Google Scholar] [CrossRef]
  87. Li, H.-J.; Pan, Y.-B.; Sun, Z.-L.; Sun, Y.-Y.; Yang, X.-T.; Feng, D.-F. Inhibition of MiR-21 Ameliorates Excessive Astrocyte Activation and Promotes Axon Regeneration Following Optic Nerve Crush. Neuropharmacology 2018, 137, 33–49. [Google Scholar] [CrossRef]
  88. Meng, F.; Henson, R.; Wehbe–Janek, H.; Ghoshal, K.; Jacob, S.T.; Patel, T. MicroRNA-21 Regulates Expression of the PTEN Tumor Suppressor Gene in Human Hepatocellular Cancer. Gastroenterology 2007, 133, 647–658. [Google Scholar] [CrossRef] [PubMed][Green Version]
  89. Kwon, Y.H.; Fingert, J.H.; Kuehn, M.H.; Alward, W.L.M. Primary Open-Angle Glaucoma. N. Engl. J. Med. 2009, 360, 1113–1124. Available online: https://www.nejm.org/doi/full/10.1056/NEJMra0804630 (accessed on 13 February 2023). [CrossRef] [PubMed][Green Version]
  90. Tabak, S.; Schreiber-Avissar, S.; Beit-Yannai, E. Crosstalk between MicroRNA and Oxidative Stress in Primary Open-Angle Glaucoma. Int. J. Mol. Sci. 2021, 22, 2421. [Google Scholar] [CrossRef]
  91. Li, Y.; Zheng, J.; Wang, X.; Wang, X.; Liu, W.; Gao, J. Mesenchymal Stem Cell-Derived Exosomes Protect Trabecular Meshwork from Oxidative Stress. Sci. Rep. 2021, 11, 14863. [Google Scholar] [CrossRef]
  92. Bradley, J.; Vranka, J.; Colvis, C.; Conger, D.; Alexander, J.; Fisk, A.; Samples, J.; Acott, T. Effect of Matrix Metalloproteinases Activity on Outflow in Perfused Human Organ Culture. Investig. Ophthalmol. Vis. Sci. 1999, 39, 2649–2658. [Google Scholar]
  93. Tamkovich, S.; Grigor’eva, A.; Eremina, A.; Tupikin, A.; Kabilov, M.; Chernykh, V.; Vlassov, V.; Ryabchikova, E. What Information Can Be Obtained from the Tears of a Patient with Primary Open Angle Glaucoma? Clin. Chim. Acta 2019, 495, 529–537. [Google Scholar] [CrossRef] [PubMed]
  94. Pantalon, A.; Obadă, O.; Constantinescu, D.; Feraru, C.; Chiseliţă, D. Inflammatory Model in Patients with Primary Open Angle Glaucoma and Diabetes. Int. J. Ophthalmol. 2019, 12, 795–801. [Google Scholar] [CrossRef] [PubMed]
  95. Li, J.; Zhou, Y.; Long, Q. Effects of Mesenchymal Stem Cells Derived Exosomes on Ultrastructure of Corneal Epithelium and Function of the Tear Film in Dry Eye BALB/c Mice. Investing. Opthalmol. Vis. Sci. 2019, 60, 4187. Available online: https://iovs.arvojournals.org/article.aspx?articleid=2743824 (accessed on 13 February 2023).
  96. Cuenca, N.; Fernández-Sánchez, L.; Campello, L.; Maneu, V.; De la Villa, P.; Lax, P.; Pinilla, I. Cellular Responses Following Retinal Injuries and Therapeutic Approaches for Neurodegenerative Diseases. Prog. Retin. Eye Res. 2014, 43, 17–75. [Google Scholar] [CrossRef] [PubMed]
  97. Deng, C.-L.; Hu, C.-B.; Ling, S.-T.; Zhao, N.; Bao, L.-H.; Zhou, F.; Xiong, Y.-C.; Chen, T.; Sui, B.-D.; Yu, X.-R.; et al. Photoreceptor Protection by Mesenchymal Stem Cell Transplantation Identifies Exosomal MiR-21 as a Therapeutic for Retinal Degeneration. Cell Death Differ. 2021, 28, 1041–1061. [Google Scholar] [CrossRef]
  98. Zhang, J.; Li, P.; Zhao, G.; He, S.; Xu, D.; Jiang, W.; Peng, Q.; Li, Z.; Xie, Z.; Zhang, H.; et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles Protect Retina in a Mouse Model of Retinitis Pigmentosa by Anti-Inflammation through MiR-146a-Nr4a3 Axis. Stem Cell Res. Ther. 2022, 13, 394. [Google Scholar] [CrossRef]
  99. Safwat, A.; Sabry, D.; Ragiae, A.; Amer, E.; Mahmoud, R.H.; Shamardan, R.M. Adipose mesenchymal stem cells–derived exosomes attenuate retina degeneration of streptozotocin-induced diabetes in rabbits. J. Circ. Biomark. 2018, 7, 1849454418807827. [Google Scholar] [CrossRef] [PubMed][Green Version]
  100. Li, W.; Jin, L.; Cui, Y.; Nie, A.; Xie, N.; Liang, G. Bone Marrow Mesenchymal Stem Cells-Induced Exosomal MicroRNA-486-3p Protects against Diabetic Retinopathy through TLR4/NF-ΚB Axis Repression. J. Endocrinol. Investig. 2021, 44, 1193–1207. [Google Scholar] [CrossRef]
  101. Li, W.; Jin, L.; Cui, Y.; Xie, N. Human Umbilical Cord Mesenchymal Stem Cells-Derived Exosomal MicroRNA-17-3p Ameliorates Inflammatory Reaction and Antioxidant Injury of Mice with Diabetic Retinopathy via Targeting STAT1. Int. Immunopharmacol. 2021, 90, 107010. [Google Scholar] [CrossRef]
  102. Gu, C.; Zhang, H.; Zhao, S.; He, D.; Gao, Y. Mesenchymal Stem Cell Exosomal MiR-146a Mediates the Regulation of the TLR4/MyD88/NF-ΚB Signaling Pathway in Inflammation Due to Diabetic Retinopathy. Comput. Math. Methods Med. 2022, 2022, 3864863. [Google Scholar] [CrossRef]
  103. Ebrahim, N.; El-Halim, H.E.A.; Helal, O.K.; El-Azab, N.E.-E.; Badr, O.A.M.; Hassouna, A.; Saihati, H.A.A.; Aborayah, N.H.; Emam, H.T.; El-wakeel, H.S.; et al. Effect of Bone Marrow Mesenchymal Stem Cells-Derived Exosomes on Diabetes-Induced Retinal Injury: Implication of Wnt/b-Catenin Signaling Pathway. Biomed. Pharmacother. 2022, 154, 113554. [Google Scholar] [CrossRef] [PubMed]
  104. Cao, X.; Xue, L.-D.; Di, Y.; Li, T.; Tian, Y.-J.; Song, Y. MSC-Derived Exosomal LncRNA SNHG7 Suppresses Endothelial-Mesenchymal Transition and Tube Formation in Diabetic Retinopathy via MiR-34a-5p/XBP1 Axis. Life Sci. 2021, 272, 119232. [Google Scholar] [CrossRef] [PubMed]
  105. Hajrasouliha, A.R.; Jiang, G.; Lu, Q.; Lu, H.; Kaplan, H.J.; Zhang, H.-G.; Shao, H. Exosomes from Retinal Astrocytes Contain Antiangiogenic Components That Inhibit Laser-Induced Choroidal Neovascularization. J. Biol. Chem. 2013, 288, 28058–28067. [Google Scholar] [CrossRef] [PubMed][Green Version]
  106. He, G.-H.; Zhang, W.; Ma, Y.-X.; Yang, J.; Chen, L.; Song, J.; Chen, S. Mesenchymal Stem Cells-Derived Exosomes Ameliorate Blue Light Stimulation in Retinal Pigment Epithelium Cells and Retinal Laser Injury by VEGF-Dependent Mechanism. Int. J. Ophthalmol. 2018, 11, 559–566. [Google Scholar] [CrossRef]
  107. Mathew, B.; Ravindran, S.; Liu, X.; Torres, L.; Chennakesavalu, M.; Huang, C.-C.; Feng, L.; Zelka, R.; Lopez, J.; Sharma, M.; et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles and Retinal Ischemia-Reperfusion. Biomaterials 2019, 197, 146–160. [Google Scholar] [CrossRef] [PubMed]
  108. Yu, Z.; Wen, Y.; Jiang, N.; Li, Z.; Guan, J.; Zhang, Y.; Deng, C.; Zhao, L.; Zheng, S.G.; Zhu, Y.; 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. [Google Scholar] [CrossRef]
  109. Ma, M.; Li, B.; Zhang, M.; Zhou, L.; Yang, F.; Ma, F.; Shao, H.; Li, Q.; Li, X.; Zhang, X. Therapeutic Effects of Mesenchymal Stem Cell-Derived Exosomes on Retinal Detachment. Exp. Eye Res. 2020, 191, 107899. [Google Scholar] [CrossRef]
  110. Dervenis, N.; Dervenis, P.; Sandinha, T.; Murphy, D.C.; Steel, D.H. Intraocular Tamponade Choice with Vitrectomy and Internal Limiting Membrane Peeling for Idiopathic Macular Hole: A Systematic Review and Meta-Analysis. Ophthalmol. Retina 2022, 6, 457–468. [Google Scholar] [CrossRef]
  111. Muqit, M.M.K.; Hamilton, R.; Ho, J.; Tucker, S.; Buck, H. Intravitreal Ocriplasmin for the Treatment of Vitreomacular Traction and Macular Hole- A Study of Efficacy and Safety Based on NICE Guidance. PLoS ONE 2018, 13, e0197072. [Google Scholar] [CrossRef][Green Version]
  112. Zhang, X.; Liu, J.; Yu, B.; Ma, F.; Ren, X.; Li, X. Effects of Mesenchymal Stem Cells and Their Exosomes on the Healing of Large and Refractory Macular Holes. Graefes Arch. Clin. Exp. Ophthalmol. 2018, 256, 2041–2052. [Google Scholar] [CrossRef]
  113. Valdes, L.M.; Sobrin, L. Uveitis Therapy: The Corticosteroid Options. Drugs 2020, 80, 765–773. [Google Scholar] [CrossRef] [PubMed]
  114. Duplechain, A.; Conrady, C.D.; Patel, B.C.; Baker, S. Uveitis. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  115. Shigemoto-Kuroda, T.; Oh, J.Y.; Kim, D.; Jeong, H.J.; Park, S.Y.; Lee, H.J.; Park, J.W.; Kim, T.W.; An, S.Y.; Prockop, D.J.; et al. MSC-Derived Extracellular Vesicles Attenuate Immune Responses in Two Autoimmune Murine Models: Type 1 Diabetes and Uveoretinitis. Stem Cell Rep. 2017, 8, 1214–1225. [Google Scholar] [CrossRef] [PubMed][Green Version]
  116. Bai, L.; Shao, H.; Wang, H.; Zhang, Z.; Su, C.; Dong, L.; Yu, B.; Chen, X.; Li, X.; Zhang, X. Effects of Mesenchymal Stem Cell-Derived Exosomes on Experimental Autoimmune Uveitis. Sci. Rep. 2017, 7, 4323. [Google Scholar] [CrossRef][Green Version]
  117. Xie, R.; Bai, L.; Yang, J.; Li, Y.; Dong, L.; Ma, F.; Li, X.; Zhang, X. Effects of rat mesenchymal stem cell-derived exosomes on rat experimental autoimmune uveitis. Chin. J. Ocul. Fundus Dis. 2018, 34, 562–567. [Google Scholar]
  118. Li, Y.; Ren, X.; Zhang, Z.; Duan, Y.; Li, H.; Chen, S.; Shao, H.; Li, X.; Zhang, X. Effect of Small Extracellular Vesicles Derived from IL-10-Overexpressing Mesenchymal Stem Cells on Experimental Autoimmune Uveitis. Stem Cell Res. Ther. 2022, 13, 100. [Google Scholar] [CrossRef]
  119. Liu, Y.; Zhou, T.; Yang, Z.; Sun, X.; Huang, Z.; Deng, X.; He, C.; Liu, X. Bone Marrow Mesenchymal Stem Cells-Derived Exosomes Attenuate Neuroinflammation and Promote Survival of Photoreceptor in Retinitis Pigmentosa. Investig. Ophthalmol. Vis. Sci. 2019, 60, 3108. [Google Scholar]
  120. Li, D.; Zhang, J.; Liu, Z.; Gong, Y.; Zheng, Z. Human Umbilical Cord Mesenchymal Stem Cell-Derived Exosomal MiR-27b Attenuates Subretinal Fibrosis via Suppressing Epithelial–Mesenchymal Transition by Targeting HOXC6. Stem Cell Res. Ther. 2021, 12, 24. [Google Scholar] [CrossRef] [PubMed]
  121. Oh, J.Y.; Kim, T.W.; Jeong, H.J.; Lee, H.J.; Ryu, J.S.; Wee, W.R.; Heo, J.W.; Kim, M.K. Intraperitoneal Infusion of Mesenchymal Stem/Stromal Cells Prevents Experimental Autoimmune Uveitis in Mice. Mediat. Inflamm. 2014, 2014, 624640. [Google Scholar] [CrossRef] [PubMed][Green Version]
  122. Li, H.; Zhang, Z.; Li, Y.; Su, L.; Duan, Y.; Zhang, H.; An, J.; Ni, T.; Li, X.; Zhang, X. Therapeutic Effect of Rapamycin-Loaded Small Extracellular Vesicles Derived from Mesenchymal Stem Cells on Experimental Autoimmune Uveitis. Front. Immunol. 2022, 13, 864956. [Google Scholar] [CrossRef]
  123. Wei, W.; Ao, Q.; Wang, X.; Cao, Y.; Liu, Y.; Zheng, S.G.; Tian, X. Mesenchymal Stem Cell–Derived Exosomes: A Promising Biological Tool in Nanomedicine. Front. Pharmacol. 2021, 11, 590470. [Google Scholar] [CrossRef]
  124. Hoang, D.M.; Pham, P.T.; Bach, T.Q.; Ngo, A.T.L.; Nguyen, Q.T.; Phan, T.T.K.; Nguyen, G.H.; Le, P.T.T.; Hoang, V.T.; Forsyth, N.R.; et al. Stem Cell-Based Therapy for Human Diseases. Signal Transduct. Target. Ther. 2022, 7, 272. [Google Scholar] [CrossRef] [PubMed]
  125. Kou, M.; Huang, L.; Yang, J.; Chiang, Z.; Chen, S.; Liu, J.; Guo, L.; Zhang, X.; Zhou, X.; Xu, X.; et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles for Immunomodulation and Regeneration: A next Generation Therapeutic Tool? Cell Death Dis. 2022, 13, 580. [Google Scholar] [CrossRef] [PubMed]
  126. Varderidou-Minasian, S.; Lorenowicz, M.J. Mesenchymal Stromal/Stem Cell-Derived Extracellular Vesicles in Tissue Repair: Challenges and Opportunities. Theranostics 2020, 10, 5979–5997. [Google Scholar] [CrossRef] [PubMed]
  127. Chen, S.; Sun, F.; Qian, H.; Xu, W.; Jiang, J. Preconditioning and Engineering Strategies for Improving the Efficacy of Mesenchymal Stem Cell-Derived Exosomes in Cell-Free Therapy. Stem Cells Int. 2022, 2022, 1779346. [Google Scholar] [CrossRef]
  128. Qazi, T.H.; Mooney, D.J.; Duda, G.N.; Geissler, S. Biomaterials That Promote Cell-Cell Interactions Enhance the Paracrine Function of MSCs. Biomaterials 2017, 140, 103–114. [Google Scholar] [CrossRef]
  129. Su, N.; Gao, P.-L.; Wang, K.; Wang, J.-Y.; Zhong, Y.; Luo, Y. Fibrous Scaffolds Potentiate the Paracrine Function of Mesenchymal Stem Cells: A New Dimension in Cell-Material Interaction. Biomaterials 2017, 141, 74–85. [Google Scholar] [CrossRef]
  130. Samsonraj, R.M.; Rai, B.; Sathiyanathan, P.; Puan, K.J.; Rötzschke, O.; Hui, J.H.; Raghunath, M.; Stanton, L.W.; Nurcombe, V.; Cool, S.M. Establishing Criteria for Human Mesenchymal Stem Cell Potency. Stem Cells 2015, 33, 1878–1891. [Google Scholar] [CrossRef]
  131. Sathiyanathan, P.; Samsonraj, R.M.; Tan, C.L.L.; Ling, L.; Lezhava, A.; Nurcombe, V.; Stanton, L.W.; Cool, S.M. A Genomic Biomarker That Identifies Human Bone Marrow-Derived Mesenchymal Stem Cells with High Scalability. Stem Cells Dayt. Ohio 2020, 38, 1124–1136. [Google Scholar] [CrossRef]
  132. Boulestreau, J.; Maumus, M.; Rozier, P.; Jorgensen, C.; Noël, D. Mesenchymal Stem Cell Derived Extracellular Vesicles in Aging. Front. Cell Dev. Biol. 2020, 8, 107. [Google Scholar] [CrossRef][Green Version]
  133. Li, Y.; Wu, Q.; Wang, Y.; Li, L.; Bu, H.; Bao, J. Senescence of Mesenchymal Stem Cells (Review). Int. J. Mol. Med. 2017, 39, 775–782. [Google Scholar] [CrossRef][Green Version]
  134. Kouroupis, D.; Churchman, S.M.; McGonagle, D.; Jones, E.A. The Assessment of CD146-Based Cell Sorting and Telomere Length Analysis for Establishing the Identity of Mesenchymal Stem Cells in Human Umbilical Cord. F1000Research 2014, 3, 126. [Google Scholar] [CrossRef] [PubMed]
  135. Laschober, G.T.; Brunauer, R.; Jamnig, A.; Fehrer, C.; Greiderer, B.; Lepperdinger, G. Leptin Receptor/CD295 Is Upregulated on Primary Human Mesenchymal Stem Cells of Advancing Biological Age and Distinctly Marks the Subpopulation of Dying Cells. Exp. Gerontol. 2009, 44, 57–62. [Google Scholar] [CrossRef] [PubMed]
  136. Jung, E.M.; Kwon, O.; Kwon, K.-S.; Cho, Y.S.; Rhee, S.K.; Min, J.-K.; Oh, D.-B. Evidences for Correlation between the Reduced VCAM-1 Expression and Hyaluronan Synthesis during Cellular Senescence of Human Mesenchymal Stem Cells. Biochem. Biophys. Res. Commun. 2011, 404, 463–469. [Google Scholar] [CrossRef]
  137. Simmons, P.J.; Torok-Storb, B. Identification of Stromal Cell Precursors in Human Bone Marrow by a Novel Monoclonal Antibody, STRO-1. Blood 1991, 78, 55–62. [Google Scholar] [CrossRef][Green Version]
  138. Yun, S.P.; Han, Y.-S.; Lee, J.H.; Kim, S.M.; Lee, S.H. Melatonin Rescues Mesenchymal Stem Cells from Senescence Induced by the Uremic Toxin p-Cresol via Inhibiting MTOR-Dependent Autophagy. Biomol. Ther. 2018, 26, 389–398. [Google Scholar] [CrossRef] [PubMed]
  139. Chaker, D.; Mouawad, C.; Azar, A.; Quilliot, D.; Achkar, I.; Fajloun, Z.; Makdissy, N. Inhibition of the RhoGTPase Cdc42 by ML141 Enhances Hepatocyte Differentiation from Human Adipose-Derived Mesenchymal Stem Cells via the Wnt5a/PI3K/MiR-122 Pathway: Impact of the Age of the Donor. Stem Cell Res. Ther. 2018, 9, 167. [Google Scholar] [CrossRef] [PubMed][Green Version]
  140. Siegel, G.; Kluba, T.; Hermanutz-Klein, U.; Bieback, K.; Northoff, H.; Schäfer, R. Phenotype, Donor Age and Gender Affect Function of Human Bone Marrow-Derived Mesenchymal Stromal Cells. BMC Med. 2013, 11, 146. [Google Scholar] [CrossRef][Green Version]
  141. Ulum, B.; Teker, H.T.; Sarikaya, A.; Balta, G.; Kuskonmaz, B.; Uckan-Cetinkaya, D.; Aerts-Kaya, F. Bone Marrow Mesenchymal Stem Cell Donors with a High Body Mass Index Display Elevated Endoplasmic Reticulum Stress and Are Functionally Impaired. J. Cell. Physiol. 2018, 233, 8429–8436. [Google Scholar] [CrossRef]
  142. Li, C.; Zhao, H.; Cheng, L.; Wang, B. Allogeneic vs. Autologous Mesenchymal Stem/Stromal Cells in Their Medication Practice. Cell Biosci. 2021, 11, 187. [Google Scholar] [CrossRef]
More
This entry is offline, you can click here to edit this entry!
Video Production Service