DOI QR코드

DOI QR Code

Umbilical cord mesenchymal stem cells derived extracellular vesicles can safely ameliorate the progression of chronic kidney diseases

  • Nassar, Wael (Department of Internal Medicine, Nephrology Section, Sahel Teaching Hospital, General Organization of Teaching Hospitals and Institutes (GOTHI)) ;
  • El-Ansary, Mervat (Department of clinical pathology, stem cells Section, Faculty of medicine, Cairo University) ;
  • Sabry, Dina (Department of Biochemistry, Faculty of medicine, Cairo University) ;
  • Mostafa, Mostafa A. (Department of Internal medicine, Nephrology Section, Faculty of Medicine, October Six University) ;
  • Fayad, Tarek (Department of Internal Medicine, Nephrology Section, Faculty of medicine, Cairo University) ;
  • Kotb, Esam (Department of Internal medicine, Nephrology Section, Faculty of Medicine, October Six University) ;
  • Temraz, Mahmoud (Department of Internal Medicine, Nephrology Section, Sahel Teaching Hospital, General Organization of Teaching Hospitals and Institutes (GOTHI)) ;
  • Saad, Abdel-Naser (Department of Internal Medicine, Nephrology Section, Sahel Teaching Hospital, General Organization of Teaching Hospitals and Institutes (GOTHI)) ;
  • Essa, Wael (Department of Internal Medicine, Nephrology Section, Sahel Teaching Hospital, General Organization of Teaching Hospitals and Institutes (GOTHI)) ;
  • Adel, Heba (Department of clinical pathology, stem cells Section, Faculty of medicine, Cairo University)
  • 투고 : 2016.04.05
  • 심사 : 2016.07.04
  • 발행 : 2016.09.01

초록

Background: Bio-products from stem/progenitor cells, such as extracellular vesicles, are likely a new promising approach for reprogramming resident cells in both acute and chronic kidney disease. Forty CKD patients stage III and IV (eGFR 15-60 mg/ml) have been divided into two groups; twenty patients as treatment group "A" and twenty patients as a matching placebo group "B". Two doses of MSC-derived extracellular vesicles had been administered to patients of group "A". Blood urea, serum creatinine, urinary albumin creatinine ratio (UACR) and estimated glomerular filtration rate (eGFR) have been used to assess kidney functions and TNF-${\alpha}$, TGF-${\beta}1$ and IL-10 have been used to assess the amelioration of the inflammatory immune activity. Results: Participants in group A exhibited significant improvement of eGFR, serum creatinine level, blood urea and UACR. Patients of the treatment group "A" also exhibited significant increase in plasma levels of TGF-${\beta}1$, and IL-10 and significant decrease in plasma levels of TNF-${\alpha}$. Participants of the control group B did not show significant improvement in any of the previously mentioned parameters at any time point of the study period. Conclusion: Administration of cell-free cord-blood mesenchymal stem cells derived extracellular vesicles (CF-CB-MSCs-EVs) is safe and can ameliorate the inflammatory immune reaction and improve the overall kidney function in grade III-IV CKD patients.

키워드

과제정보

연구 과제 주관 기관 : Hegaz Nephrology Center (HNC)

참고문헌

  1. Xiangyu Zou, Guangyuan Zhang, Zhongliang Cheng, Deming Yin, Tao Du, Guanqun Ju, Shuai Miao,Guohua Liu, Mujun Lu and Yingjian Zhu. Microvesicles derived from human Wharton's Jelly mesenchymal stromal cells ameliorate renal ischemia-reperfusion injury in rats by suppressing CX3CL1. Stem Cell Research & Therapy. 2014;5:40. https://doi.org/10.1186/scrt428
  2. Ronne Wee Yeh Y, Ruenn Chai L, Kok Hian T, Sai Kiang L. Exosome: a novel and safer therapeutic refinement of mesenchymal stem cell. Exosomes microvesicles. 2013;1:7.
  3. Yu B, Zhang X, Li X. Exosomes derived from mesenchymal stem cells. Int J Mol Sci. 2014;15:4142-57. https://doi.org/10.3390/ijms15034142
  4. Camussi G, Deregibus MC, Bruno S, Cantaluppi V, Biancone L. Exosomes/microvesicles as a mechanism of cell-to-cell communication. Kidney Int. 2010;78:838-48. https://doi.org/10.1038/ki.2010.278
  5. Ying WANG, Li-ming CHEN, Ming-lin LIU. Microvesicles and diabetic complications - novel mediators, potential biomarkers and therapeutic targets. Acta Pharmacologica Sinica, npg. 2014;35:433-43. https://doi.org/10.1038/aps.2013.188
  6. Chen YT, Sun CK, Lin YC, Chang LT, Chen YL, Tsai TH, Chung SY, Chua S, Kao YH, Yen CH, Shao PL, Chang KC, Leu S, Yip HK. Adipose-derived mesenchymal stem cell protects kidneys against ischemia-reperfusion injury through suppressing oxidative stress and inflammatory reaction. J Transl Med. 2011;9:51. https://doi.org/10.1186/1479-5876-9-51
  7. Yong Zhao, Zhaoshun Jiang, Tingbao Zhao, Mingliang Ye, Chengjin Hu, Zhaohui Yin, Heng Li, Ye Zhang, Yalin Diao, Yunxiang Li, Yingjian Chen, Xiaoming Sun, Mary Beth Fisk, Randal Skidgel, Mark Holterman, Bellur P and Theodore M. Reversal of type 1 diabetes via islet ${\beta}$ cell regeneration following immune modulation by cord blood-derived multipotent stem cells. BMC Medicine. 2012;10:3. 10. https://doi.org/10.1186/1741-7015-10-3
  8. Cavaglieri RC, Martini D, Sogayar MC, Noronha IL. Mesenchymal stem cells delivered at the subcapsule of the kidney ameliorate renal disease in the rat remnant kidney model. Transplant Proc. 2009;41:947-51. https://doi.org/10.1016/j.transproceed.2009.01.072
  9. Villanueva S, Ewertz E, Carrion F, Tapia A, Vergara C, Cespedes C, Saez PJ, Luz P, Irarrazabal C, Carreno JE, Figueroa F, Vio CP. Mesenchymal stem cell injection ameliorates chronic renal failure in a rat model. Clin Sci (Lond). 2011;121:489-99. https://doi.org/10.1042/CS20110108
  10. Gatti S, Bruno S, Deregibus MC, et al. Microvesicles derived from human adult mesenchymal stem cells protect against ischaemia reperfusioninduced acute and chronic kidney injury. Nephrol Dial Transplant. 2011;26:1474-83. https://doi.org/10.1093/ndt/gfr015
  11. Morigi M, Imberti B, Zoja C, Corna D, Tomasoni S, et al. Mesenchymal stem cells are renotropic, helping to repair the kidney and improve function in acute renal failure. J Am Soc Nephrol. 2004;15:1794-804. https://doi.org/10.1097/01.ASN.0000128974.07460.34
  12. Biancone L, Bruno S, Deregibus MC, Tetta C, Camussi G. Therapeutic potential of mesenchymal stem cell-derived microvesicles. Nephrol Dial Transplant. 2012;27:3037-42. https://doi.org/10.1093/ndt/gfs168
  13. Morigi M, Introna M, Imberti B, et al. Human bone marrow mesenchymal stem cells accelerate recovery of acute renal injury and prolong survival in mice. STEM CELLS. 2008;26:2075-82. https://doi.org/10.1634/stemcells.2007-0795
  14. Guidance for Industry. Biologics License Applications for Minimally Manipulated, Unrelated Allogeneic Placental/Umbilical Cord Blood Intended for Hematopoietic and Immunologic Reconstitution in Patients with Disorders Affecting the Hematopoietic System. 2014. U.S. Department of Health and Human Services, Food and Drug Administration, Center for Biologics Evaluation and Research.
  15. Herrera MB, Bussolati B, Bruno S, et al. Mesenchymal stem cells contribute to the renal repair of acute tubular epithelial injury. Int J Mol Med. 2004;14:1035-41.
  16. Aggarwal S, Moggio A, Bussolati B. Concise review: stem/progenitor cells for renal tissue repair: current knowledge and perspectives. Stem Cells Trans Med published online October. 2013. published online October 28, 2013.
  17. Oscar PB W, Nordin JZ, Aisling O'L, Ylva G, et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. Journal of Extracellular Vesicles. 2015;4:26316. https://doi.org/10.3402/jev.v4.26316
  18. Lai RC, Arslan F, Lee MM, Sze NS, Choo A, Chen TS, et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010;4:214-22. https://doi.org/10.1016/j.scr.2009.12.003
  19. Stefania B, Cristina G, Federica C, Maria Chiara D, Vincenzo C, Luigi B, Ciro T, Giovanni C. Microvesicles derived from mesenchymal stem cells enhance survival in a lethal model of acute kidney injury. PLoS One. 2012;7:3.
  20. Morigi M, Rota C, Montemurro T, et al. Life-sparing effect of human cord blood-mesenchymal stem cells in experimental acute kidney injury. STEM CELLS. 2010;28:513-22.
  21. Bruno S, Grange C, Deregibus MC, Calogero RA, Saviozzi S, Collino F, Morando L, Busca A, Falda M, Bussolati B, Tetta C, Camussi G. Mesenchymal stem cell-derived microvesicles protect against acute tubular injury. J Am Soc Nephrol. 2009;20:1053-67. https://doi.org/10.1681/ASN.2008070798
  22. Zhao Y, Mazzone T. Human cord blood stem cells and the journey to a cure for type 1 diabetes. Autoimmun Rev. 2010;10:103-7. https://doi.org/10.1016/j.autrev.2010.08.011
  23. Zhao Y, Lin B, Darflinger R, Zhang Y, Holterman MJ, Skidgel RA. Human cord blood stem cell-modulated regulatory T lymphocytes reverse the autoimmune-caused type 1 diabetes in non-obese diabetic (NOD) mice. PLoS One. 2009;4:e4226. https://doi.org/10.1371/journal.pone.0004226
  24. Bi B, Schmitt R, Israilova M, Nishio H, Cantley LG. Stromal cells protect against acute tubular injury via an endocrine effect. J Am Soc Nephrol. 2007;18:2486-96. https://doi.org/10.1681/ASN.2007020140
  25. Bruno S, Camussi G. Ro le of mesenchymal stem cell-derived microvesicles in tissue repair. Pediatr Nephrol. 2013;28(12):2249-54. https://doi.org/10.1007/s00467-013-2413-z
  26. English K, Ryan JM, Tobin L, Murphy MJ, Barry FP, Mahon BP. Cell contact, prostaglandin E(2) and transforming growth factor beta 1 play non-redundant roles in human mesenchymal stem cell induction of CD4+CD25 (High) forkhead box P3+ regulatory T cells. Clin Exp Immunol. 2009;15:149-60.
  27. Du T, Zou X, Cheng J, Wu S, Zhong L, Ju G, Zhu J, Liu G, Zhu Y, Xia S. Human Wharton's jelly-derived mesenchymal stromal cells reduce renal fibrosis through induction of native and foreign hepatocyte growth factor synthesis in injured tubular epithelial cells. Stem Cell Res Ther. 2013;4:59. https://doi.org/10.1186/scrt215
  28. Tomasoni S, Longaretti L, Rota C, Morigi M, Conti S, Gotti E, et al. Transfer of growth factor receptor mRNA via exosomes unravels the regenerative effect of mesenchymal stem cells. Stem Cells Dev. 2013;22:772-80. https://doi.org/10.1089/scd.2012.0266
  29. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosomemediated transfer of mRNAs an microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654-9. https://doi.org/10.1038/ncb1596
  30. Kusaba T, Lalli M, Kramann R, Kobayashi A, Humphreys BD. Differentiated kidney epithelial cells repair injured proximal tubule. PNAS. 2014;11(15):5753-4.

피인용 문헌

  1. Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles and Their Potential as Novel Immunomodulatory Therapeutic Agents vol.18, pp.7, 2016, https://doi.org/10.3390/ijms18071450
  2. Challenges and Strategies for Improving the Regenerative Effects of Mesenchymal Stromal Cell-Based Therapies vol.18, pp.10, 2016, https://doi.org/10.3390/ijms18102087
  3. Mesenchymal stem cell-derived extracellular vesicles for kidney repair: current status and looming challenges vol.8, pp.1, 2017, https://doi.org/10.1186/s13287-017-0727-7
  4. Erratum to: Umbilical cord mesenchymal stem cells derived extracellular vesicles can safely ameliorate the progression of chronic kidney diseases vol.21, pp.1, 2017, https://doi.org/10.1186/s40824-017-0089-3
  5. Bioprocessing of Mesenchymal Stem Cells and Their Derivatives: Toward Cell-Free Therapeutics vol.2018, pp.None, 2016, https://doi.org/10.1155/2018/9415367
  6. Extracellular Vesicles: A New Prospective in Crosstalk between Microenvironment and Stem Cells in Hematological Malignancies vol.2018, pp.None, 2016, https://doi.org/10.1155/2018/9863194
  7. Therapeutic potential of products derived from mesenchymal stem/stromal cells in pulmonary disease vol.19, pp.None, 2016, https://doi.org/10.1186/s12931-018-0921-x
  8. Immunothrombotic Activity of Damage-Associated Molecular Patterns and Extracellular Vesicles in Secondary Organ Failure Induced by Trauma and Sterile Insults vol.9, pp.None, 2016, https://doi.org/10.3389/fimmu.2018.00190
  9. Stem/Stromal Cells for Treatment of Kidney Injuries With Focus on Preclinical Models vol.5, pp.None, 2016, https://doi.org/10.3389/fmed.2018.00179
  10. Dynamic Cultivation of Mesenchymal Stem Cell Aggregates vol.5, pp.2, 2016, https://doi.org/10.3390/bioengineering5020048
  11. Regenerative medicine in kidney disease: where we stand and where to go vol.33, pp.9, 2018, https://doi.org/10.1007/s00467-017-3754-9
  12. Histological and immunohistochemical study of the potential therapeutic impacts of bone marrow mesenchymal stem cells and exosomes for sciatic nerve crush injury model in rats vol.41, pp.4, 2018, https://doi.org/10.1080/01478885.2018.1505205
  13. Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Promote Neuroprotection in a Genetic DBA/2J Mouse Model of Glaucoma vol.59, pp.13, 2018, https://doi.org/10.1167/iovs.18-25310
  14. Mesenchymal stem cell-derived extracellular vesicles attenuate influenza virus-induced acute lung injury in a pig model vol.9, pp.1, 2016, https://doi.org/10.1186/s13287-018-0774-8
  15. Effect of Adipose-Derived Stem Cells and Their Exo as Adjunctive Therapy to Nonsurgical Periodontal Treatment: A Histologic and Histomorphometric Study in Rats vol.8, pp.4, 2018, https://doi.org/10.3390/biom8040167
  16. Mesenchymal Stem Cell-Derived Exosomes Ameliorated Diabetic Nephropathy by Autophagy Induction through the mTOR Signaling Pathway vol.7, pp.12, 2016, https://doi.org/10.3390/cells7120226
  17. Effects of Extracellular Vesicles Derived from Mesenchymal Stem/Stromal Cells on Liver Diseases vol.14, pp.5, 2016, https://doi.org/10.2174/1574888x14666190308123714
  18. Emergent Prophylactic, Reparative and Restorative Brain Interventions for Infants Born Preterm With Cerebral Palsy vol.10, pp.None, 2016, https://doi.org/10.3389/fphys.2019.00015
  19. Stem Cell-Derived Extracellular Vesicles as Immunomodulatory Therapeutics vol.2019, pp.None, 2019, https://doi.org/10.1155/2019/5126156
  20. Exosome as a Novel Shuttle for Delivery of Therapeutics across Biological Barriers vol.16, pp.1, 2019, https://doi.org/10.1021/acs.molpharmaceut.8b00901
  21. Potential and Therapeutic Efficacy of Cell-based Therapy Using Mesenchymal Stem Cells for Acute/chronic Kidney Disease vol.20, pp.7, 2016, https://doi.org/10.3390/ijms20071619
  22. Extracellular vesicles in renal physiology and clinical applications for renal disease vol.34, pp.3, 2016, https://doi.org/10.3904/kjim.2019.108
  23. Advances in therapeutic applications of extracellular vesicles vol.11, pp.492, 2019, https://doi.org/10.1126/scitranslmed.aav8521
  24. Harnessing the mesenchymal stem cell secretome for regenerative urology vol.16, pp.6, 2019, https://doi.org/10.1038/s41585-019-0169-3
  25. Mesenchymal stem cell-derived exosomes for clinical use vol.54, pp.suppl2, 2019, https://doi.org/10.1038/s41409-019-0616-z
  26. A Retrospective Analysis of Safety and Efficacy of Wharton’s Jelly Stem Cell Administration in Children with Spina Bifida vol.15, pp.5, 2019, https://doi.org/10.1007/s12015-019-09902-6
  27. Stem Cell-Derived Extracellular Vesicles and Kidney Regeneration vol.8, pp.10, 2019, https://doi.org/10.3390/cells8101240
  28. Cell-derived Secretome for the Treatment of Renal Disease vol.23, pp.2, 2016, https://doi.org/10.3339/jkspn.2019.23.2.67
  29. Conditioned Medium of Mesenchymal Stromal Cells: A New Class of Therapeutics vol.84, pp.11, 2019, https://doi.org/10.1134/s0006297919110129
  30. Novel insights into MSC-EVs therapy for immune diseases vol.7, pp.None, 2019, https://doi.org/10.1186/s40364-019-0156-0
  31. Stem cell-derived extracellular vesicles for renal repair: do cardiovascular comorbidities matter? vol.317, pp.6, 2016, https://doi.org/10.1152/ajprenal.00434.2019
  32. Regenerative Role of Stem Cell-Derived Extracellular Vesicles in Acute Kidney Injury vol.144, pp.12, 2016, https://doi.org/10.1159/000511347
  33. Extracellular vesicle-based Nanotherapeutics: Emerging frontiers in anti-inflammatory therapy vol.10, pp.18, 2016, https://doi.org/10.7150/thno.47865
  34. Mesenchymal stem cell‐derived extracellular vesicles for the treatment of acute respiratory distress syndrome vol.9, pp.1, 2016, https://doi.org/10.1002/sctm.19-0205
  35. Human Breast Milk-Derived Extracellular Vesicles in the Protection Against Experimental Necrotizing Enterocolitis vol.55, pp.1, 2016, https://doi.org/10.1016/j.jpedsurg.2019.09.052
  36. Comparison of the Effects of Mesenchymal Stem Cells with Their Extracellular Vesicles on the Treatment of Kidney Damage Induced by Chronic Renal Artery Stenosis vol.2020, pp.None, 2016, https://doi.org/10.1155/2020/8814574
  37. Mesenchymal Stem Cell-Derived Extracellular Vesicles and Their Therapeutic Potential vol.2020, pp.None, 2016, https://doi.org/10.1155/2020/8825771
  38. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Opportunities and Challenges for Clinical Translation vol.8, pp.None, 2016, https://doi.org/10.3389/fbioe.2020.00997
  39. Exosomes in Nephropathies: A Rich Source of Novel Biomarkers vol.2020, pp.None, 2016, https://doi.org/10.1155/2020/8897833
  40. Nephroprotective Potential of Mesenchymal Stromal Cells and Their Extracellular Vesicles in a Murine Model of Chronic Cyclosporine Nephrotoxicity vol.8, pp.None, 2020, https://doi.org/10.3389/fcell.2020.00296
  41. Perspectives for Future Use of Extracellular Vesicles from Umbilical Cord- and Adipose Tissue-Derived Mesenchymal Stem/Stromal Cells in Regenerative Therapies—Synthetic Review vol.21, pp.3, 2016, https://doi.org/10.3390/ijms21030799
  42. The potential neuroprotective role of mesenchymal stem cell-derived exosomes in cerebellar cortex lipopolysaccharide-induced neuroinflammation in rats: a histological and immunohistochemical study vol.44, pp.2, 2016, https://doi.org/10.1080/01913123.2020.1726547
  43. The Osteosarcoma Microenvironment: A Complex but Targetable Ecosystem vol.9, pp.4, 2016, https://doi.org/10.3390/cells9040976
  44. Mesenchymal Stem Cell Derived Extracellular Vesicles for Tissue Engineering and Regenerative Medicine Applications vol.9, pp.4, 2020, https://doi.org/10.3390/cells9040991
  45. In Vivo Tracking of Mesenchymal Stem Cell-Derived Extracellular Vesicles Improving Mitochondrial Function in Renal Ischemia-Reperfusion Injury vol.14, pp.4, 2020, https://doi.org/10.1021/acsnano.9b08207
  46. Kidney Regenerative Medicine: Promises and Limitations vol.7, pp.2, 2016, https://doi.org/10.1007/s40472-020-00273-3
  47. Mesenchymal Stem Cell Derived Biocompatible Membrane Vesicles Demonstrate Immunomodulatory Activity Inhibiting Activation and proliferation of Human Mononuclear Cells vol.12, pp.6, 2016, https://doi.org/10.3390/pharmaceutics12060577
  48. Mesenchymal stem cell derived extracellular vesicles: promising immunomodulators against autoimmune, autoinflammatory disorders and SARS-CoV-2 infection vol.44, pp.3, 2016, https://doi.org/10.3906/biy-2002-79
  49. Damage-associated molecular patterns in trauma vol.46, pp.4, 2016, https://doi.org/10.1007/s00068-019-01235-w
  50. Biomaterials Functionalized with MSC Secreted Extracellular Vesicles and Soluble Factors for Tissue Regeneration vol.30, pp.37, 2016, https://doi.org/10.1002/adfm.201909125
  51. International Society for Extracellular Vesicles and International Society for Cell and Gene Therapy statement on extracellular vesicles from mesenchymal stromal cells and other cells: considerations vol.22, pp.9, 2020, https://doi.org/10.1016/j.jcyt.2020.05.002
  52. Gene Editing by Extracellular Vesicles vol.21, pp.19, 2016, https://doi.org/10.3390/ijms21197362
  53. Extracellular Vesicle-Based Nucleic Acid Delivery: Current Advances and Future Perspectives in Cancer Therapeutic Strategies vol.12, pp.10, 2016, https://doi.org/10.3390/pharmaceutics12100980
  54. Mesenchymal Stem Cell-Derived Extracellular Vesicles: A Novel Cell-Free Therapy vol.49, pp.7, 2016, https://doi.org/10.1080/08820139.2020.1712416
  55. Therapeutic effects of extracellular vesicles from human adipose‐derived mesenchymal stem cells on chronic experimental autoimmune encephalomyelitis vol.235, pp.11, 2016, https://doi.org/10.1002/jcp.29721
  56. Functional assays to assess the therapeutic potential of extracellular vesicles vol.10, pp.1, 2016, https://doi.org/10.1002/jev2.12033
  57. Combination therapy with melatonin, stem cells and extracellular vesicles is effective in limiting renal ischemia–reperfusion injury in a rat model vol.27, pp.11, 2016, https://doi.org/10.1111/iju.14345
  58. Therapeutic Potential of Extracellular Vesicles for the Treatment of Corneal Injuries and Scars vol.9, pp.12, 2016, https://doi.org/10.1167/tvst.9.12.1
  59. Mesenchymal stem cell derived-exosomes: a modern approach in translational medicine vol.18, pp.1, 2020, https://doi.org/10.1186/s12967-020-02622-3
  60. Mesenchymal stem cell-derived exosome: a promising alternative in the therapy of Alzheimer’s disease vol.12, pp.1, 2016, https://doi.org/10.1186/s13195-020-00670-x
  61. Extracellular vesicles for acute kidney injury in preclinical rodent models: a meta-analysis vol.11, pp.1, 2016, https://doi.org/10.1186/s13287-019-1530-4
  62. Three-dimensional culture of MSCs produces exosomes with improved yield and enhanced therapeutic efficacy for cisplatin-induced acute kidney injury vol.11, pp.1, 2016, https://doi.org/10.1186/s13287-020-01719-2
  63. BMMSC-sEV-derived miR-328a-3p promotes ECM remodeling of damaged urethral sphincters via the Sirt7/TGFβ signaling pathway vol.11, pp.1, 2020, https://doi.org/10.1186/s13287-020-01808-2
  64. Comparison of two ASC-derived therapeutics in an in vitro OA model: secretome versus extracellular vesicles vol.11, pp.1, 2016, https://doi.org/10.1186/s13287-020-02035-5
  65. Extracellular vesicles derived from different sources of mesenchymal stem cells: therapeutic effects and translational potential vol.10, pp.1, 2016, https://doi.org/10.1186/s13578-020-00427-x
  66. Improving hematopoietic engraftment: Potential role of mesenchymal stromal cell‐derived extracellular vesicles vol.39, pp.1, 2016, https://doi.org/10.1002/stem.3278
  67. Exosomes: A new horizon in modern medicine vol.264, pp.None, 2016, https://doi.org/10.1016/j.lfs.2020.118623
  68. Stem Cell-Derived Nanovesicles: A Novel Cell-Free Therapy for Wound Healing vol.2021, pp.None, 2016, https://doi.org/10.1155/2021/1285087
  69. Immunomodulatory and Regenerative Effects of Mesenchymal Stem Cells and Extracellular Vesicles: Therapeutic Outlook for Inflammatory and Degenerative Diseases vol.11, pp.None, 2016, https://doi.org/10.3389/fimmu.2020.591065
  70. Apoptotic endocrinal toxic effects of perchlorate in human placental cells vol.8, pp.None, 2016, https://doi.org/10.1016/j.toxrep.2021.04.002
  71. The Micro-RNA Cargo of Extracellular Vesicles Released by Human Adipose Tissue-Derived Mesenchymal Stem Cells Is Modified by Obesity vol.9, pp.None, 2016, https://doi.org/10.3389/fcell.2021.660851
  72. From Mesenchymal Stromal Cells to Engineered Extracellular Vesicles: A New Therapeutic Paradigm vol.9, pp.None, 2021, https://doi.org/10.3389/fcell.2021.705676
  73. The Role of Mesenchymal Stromal Cells-Derived Small Extracellular Vesicles in Diabetes and Its Chronic Complications vol.12, pp.None, 2021, https://doi.org/10.3389/fendo.2021.780974
  74. Application of Stem Cell-Derived Extracellular Vesicles as an Innovative Theranostics in Microbial Diseases vol.12, pp.None, 2021, https://doi.org/10.3389/fmicb.2021.785856
  75. Overview and Update on Methods for Cargo Loading into Extracellular Vesicles vol.9, pp.2, 2016, https://doi.org/10.3390/pr9020356
  76. Extracellular vesicles from organoids and 3D culture systems vol.118, pp.3, 2016, https://doi.org/10.1002/bit.27606
  77. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Regenerative Potential and Challenges vol.10, pp.3, 2021, https://doi.org/10.3390/biology10030172
  78. Novel Diagnostics and Therapeutics in Sepsis vol.9, pp.3, 2016, https://doi.org/10.3390/biomedicines9030311
  79. Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles Elicit Better Preservation of the Intra-Renal Microvasculature Than Renal Revascularization in Pigs with Renovascular Disease vol.10, pp.4, 2016, https://doi.org/10.3390/cells10040763
  80. Critical considerations for the development of potency tests for therapeutic applications of mesenchymal stromal cell-derived small extracellular vesicles vol.23, pp.5, 2016, https://doi.org/10.1016/j.jcyt.2021.01.001
  81. Exploring interactions between extracellular vesicles and cells for innovative drug delivery system design vol.173, pp.None, 2021, https://doi.org/10.1016/j.addr.2021.03.017
  82. Extracellular Vesicles as a Therapeutic Tool for Kidney Disease: Current Advances and Perspectives vol.22, pp.11, 2016, https://doi.org/10.3390/ijms22115787
  83. Endothelial Progenitor Cell-Derived Extracellular Vesicles: Potential Therapeutic Application in Tissue Repair and Regeneration vol.22, pp.12, 2021, https://doi.org/10.3390/ijms22126375
  84. Mesenchymal Stem Cell-Derived Extracellular Vesicles to the Rescue of Renal Injury vol.22, pp.12, 2016, https://doi.org/10.3390/ijms22126596
  85. Camouflage strategies for therapeutic exosomes evasion from phagocytosis vol.31, pp.None, 2021, https://doi.org/10.1016/j.jare.2021.01.001
  86. Comparative study of commercial protocols for high recovery of high-purity mesenchymal stem cell-derived extracellular vesicle isolation and their efficient labeling with fluorescent dyes vol.35, pp.None, 2021, https://doi.org/10.1016/j.nano.2021.102396
  87. Umbilical Cord Mesenchymal Stromal Cell-Derived Exosomes Rescue the Loss of Outer Hair Cells and Repair Cochlear Damage in Cisplatin-Injected Mice vol.22, pp.13, 2021, https://doi.org/10.3390/ijms22136664
  88. Extracellular vesicles as a next-generation drug delivery platform vol.16, pp.7, 2016, https://doi.org/10.1038/s41565-021-00931-2
  89. Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles for Chronic Kidney Disease: Are We There Yet? vol.78, pp.2, 2016, https://doi.org/10.1161/hypertensionaha.121.14596
  90. High Therapeutic and Esthetic Properties of Extracellular Vesicles Produced from the Stem Cells and Their Spheroids Cultured from Ocular Surgery-Derived Waste Orbicularis Oculi Muscle Tissues vol.10, pp.8, 2021, https://doi.org/10.3390/antiox10081292
  91. Extracellular Vesicles in Blood: Sources, Effects, and Applications vol.22, pp.15, 2016, https://doi.org/10.3390/ijms22158163
  92. Mesenchymal Stem Cell-Derived Exosomes as an Emerging Paradigm for Regenerative Therapy and Nano-Medicine: A Comprehensive Review vol.11, pp.8, 2016, https://doi.org/10.3390/life11080784
  93. The role of extracellular vesicles in podocyte autophagy in kidney disease vol.15, pp.3, 2021, https://doi.org/10.1007/s12079-020-00594-z
  94. TNF-α and IFN-γ Participate in Improving the Immunoregulatory Capacity of Mesenchymal Stem/Stromal Cells: Importance of Cell-Cell Contact and Extracellular Vesicles vol.22, pp.17, 2016, https://doi.org/10.3390/ijms22179531
  95. Therapeutic Implications of Mesenchymal Stromal Cells and Their Extracellular Vesicles in Autoimmune Diseases: From Biology to Clinical Applications vol.22, pp.18, 2016, https://doi.org/10.3390/ijms221810132
  96. Exosomes as mediators of intercellular crosstalk in metabolism vol.33, pp.9, 2021, https://doi.org/10.1016/j.cmet.2021.08.006
  97. Extracellular Vesicles in Human Milk vol.14, pp.10, 2021, https://doi.org/10.3390/ph14101050
  98. A brief history of nearly EV‐erything - The rise and rise of extracellular vesicles vol.10, pp.14, 2016, https://doi.org/10.1002/jev2.12144
  99. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles attenuate experimental autoimmune encephalomyelitis via regulating pro and anti-inflammatory cytokines vol.11, pp.1, 2016, https://doi.org/10.1038/s41598-021-91291-3
  100. Coassembly of hypoxia-sensitive macrocyclic amphiphiles and extracellular vesicles for targeted kidney injury imaging and therapy vol.19, pp.1, 2016, https://doi.org/10.1186/s12951-021-01192-w
  101. Stem cells and exosomes: promising candidates for necrotizing enterocolitis therapy vol.12, pp.1, 2016, https://doi.org/10.1186/s13287-021-02389-4
  102. Immunomodulatory extracellular vesicles: an alternative to cell therapy for COVID-19 vol.21, pp.12, 2021, https://doi.org/10.1080/14712598.2021.1921141
  103. Effects of Adipose-Derived Biogenic Nanoparticle-Associated microRNA-451a on Toll-like Receptor 4-Induced Cytokines vol.14, pp.1, 2016, https://doi.org/10.3390/pharmaceutics14010016