DOI QR코드

DOI QR Code

Multiplexed targeting of microRNA in stem cell-derived extracellular vesicles for regenerative medicine

  • Song, Byeong-Wook (Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary's Hospital) ;
  • Oh, Sekyung (Department of Medical Science, College of Medicine, Catholic Kwandong University) ;
  • Chang, Woochul (Department of Biology Education, College of Education, Pusan National University)
  • 투고 : 2021.11.23
  • 심사 : 2022.01.10
  • 발행 : 2022.02.28

초록

Regenerative medicine is a research field that develops methods to restore damaged cell or tissue function by regeneration, repair or replacement. Stem cells are the raw material of the body that is ultimately used from the point of view of regenerative medicine, and stem cell therapy uses cells themselves or their derivatives to promote responses to diseases and dysfunctions, the ultimate goal of regenerative medicine. Stem cell-derived extracellular vesicles (EVs) are recognized as an attractive source because they can enrich exogenous microRNAs (miRNAs) by targeting pathological recipient cells for disease therapy and can overcome the obstacles faced by current cell therapy agents. However, there are some limitations that need to be addressed before using miRNA-enriched EVs derived from stem cells for multiplexed therapeutic targeting in many diseases. Here, we review various roles on miRNA-based stem cell EVs that can induce effective and stable functional improvement of stem cell-derived EVs. In addition, we introduce and review the implications of several miRNA-enriched EV therapies improved by multiplexed targeting in diseases involving the circulatory system and nervous system. This systemic review may offer potential roles for stem cell-derived therapeutics with multiplexed targeting.

키워드

과제정보

This work was supported by a 2-Year Research Grant of Pusan National University.

참고문헌

  1. Mao AS and Mooney DJ (2015) Regenerative medicine: Current therapies and future directions. Proc Natl Acad Sci U S A 112, 14452-14459 https://doi.org/10.1073/pnas.1508520112
  2. Hu C, Zhao L, Zhang L, Bao Q and Li L (2020) Mesenchymal stem cell-based cell-free strategies: safe and effective treatments for liver injury. Stem Cell Res Ther 11, 377 https://doi.org/10.1186/s13287-020-01895-1
  3. Stoltz JF, de Isla N, Li YP et al (2015) Stem cells and regenerative medicine: myth or reality of the 21th century. Stem Cells Int 2015, 734731 https://doi.org/10.1155/2015/734731
  4. Song BW and Hwang KC (2019) Developmental strategy of stem cell therapy for improving clinically-hostile environment. Ann Stem Cell Res Ther 3, 1033
  5. Salazar-Noratto GE, Luo G, Denoeud C et al (2019) Understanding and leveraging cell metabolism to enhance mesenchymal stem cell transplantation survival in tissue engineering and regenerative medicine applications. Stem Cells 38, 22-33 https://doi.org/10.1002/stem.3079
  6. Kurtz A (2008) Mesenchymal stem cell delivery routes and fate. Int J Stem Cells 1, 1-7 https://doi.org/10.15283/ijsc.2008.1.1.1
  7. Volarevic V, Markovic BS, Gazdic M et al (2018) Ethical and safety issues of stem cell-based therapy. Int J Med Sci 15, 36-45 https://doi.org/10.7150/ijms.21666
  8. Prockop DJ, Brenner M, Fibbe WE et al (2010) Defining the risks of mesenchymal stromal cell therapy. Cytotherapy 12, 576-578 https://doi.org/10.3109/14653249.2010.507330
  9. Toma C, Pittenger MF, Cahill KS, Byrne BJ and Kessler PD (2002) Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation 105, 93-98 https://doi.org/10.1161/hc0102.101442
  10. McGinley LM, McMahon J, Stocca A et al (2013) Mesenchymal stem cell survival in the infarcted heart is enhanced by lentivirus vector-mediated heat shock protein 27 expression. Hum Gene Ther 24, 840-851 https://doi.org/10.1089/hum.2011.009
  11. Chia YC, Anjum CE, Yee HR et al (2020) Stem cell therapy for neurodegenerative diseases: how do stem cells bypass the blood-brain barrier and home to the brain? Stem Cells Int 2020, 8889061
  12. Baglio SR, Pegtel DM and Baldini N (2012) Mesenchymal stem cell secreted vesicles provide novel opportunities in (stem) cell-free therapy. Front Physiol 6, 359 https://doi.org/10.3389/fphys.2015.00359
  13. Makarova J, Turchinovich A, Shkurnikov M and Tonevitsky A (2021) Extracellular miRNAs and cell-cell communication: problems and prospects. Trends Biochem Sci 46, 640-651 https://doi.org/10.1016/j.tibs.2021.01.007
  14. Song BW, Lee CY, Kim R et al (2021) Multiplexed targeting of miRNA-210 in stem cell-derived extracellular vesicles promotes selective regeneration in ischemic hearts. Exp Mol Med 53, 695-708 https://doi.org/10.1038/s12276-021-00584-0
  15. Thery C, Witwer KW, Aikawa et al (2018) Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 7, 1535750 https://doi.org/10.1080/20013078.2018.1535750
  16. Crescitelli R, Lasser C, Szabo TG et al (2013) Distinct RNA profiles in subpopulations of extracellular vesicles: apoptotic bodies, microvesicles and exosomes. J Extracell Vesicles 2, 20677 https://doi.org/10.3402/jev.v2i0.20677
  17. Jeppesen DK, Fenix AM, Franklin JL et al (2019) Reassessment of exosome composition. Cell 177, 428-445.e18 https://doi.org/10.1016/j.cell.2019.02.029
  18. Doyle LM and Wang MZ (2019) Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells 8, 727 https://doi.org/10.3390/cells8070727
  19. O'Brien K, Breyne K, Ughetto S, Laurent LC and Breakefield XO (2020) RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat Rev Mol Cell Biol 21, 585-606 https://doi.org/10.1038/s41580-020-0251-y
  20. Kim HS, Choi DY, Yun SJ et al (2012) Proteomic analysis of microvesicles derived from human mesenchymal stem cells. J Proteome Res 11, 839-849 https://doi.org/10.1021/pr200682z
  21. Lai RC, Tan SS, Teh BJ et al (2012) Proteolytic potential of the MSC exosome proteome: implications for an exosome-mediated delivery of therapeutic proteasome. Int J Proteomics 2012, 971907 https://doi.org/10.1155/2012/971907
  22. Wei Z, Batagov AO, Schinelli S et al (2017) Coding and noncoding landscape of extracellular RNA released by human glioma stem cells. Nat Commun 8, 1145 https://doi.org/10.1038/s41467-017-01196-x
  23. de la Cuesta F, Passalacqua I, Rodor J, Bhushan R, Denby L and Baker AH (2019) Extracellular vesicle cross-talk between pulmonary artery smooth muscle cells and endothelium during excessive TGF-β signalling: implications for PAH vascular remodelling. Cell Commun Signal 17, 143 https://doi.org/10.1186/s12964-019-0449-9
  24. Chen L, Wang Y, Li S et al (2020) Exosomes derived from GDNF-modified human adipose mesenchymal stem cells ameliorate peritubular capillary loss in tubulointerstitial fibrosis by activating the SIRT1/eNOS signaling pathway. Theranostics 10, 9425-9442 https://doi.org/10.7150/thno.43315
  25. Zhang X, Jiang Y, Huang Q et al (2021) Exosomes derived from adipose-derived stem cells overexpressing glyoxalase-1 protect endothelial cells and enhance angiogenesis in type 2 diabetic mice with limb ischemia. Stem Cell Res Ther 12, 403 https://doi.org/10.1186/s13287-021-02475-7
  26. Dykxhoorn DM, Novina CD and Sharp PA (2003) Killing the messenger: short RNAs that silence gene expression. Nat Rev Mol Cell Biol 4, 457-467 https://doi.org/10.1038/nrm1129
  27. Kim D, Chang HR and Baek D (2017) Rules for functional microRNA targeting. BMB Rep 50, 554-559 https://doi.org/10.5483/BMBRep.2017.50.11.179
  28. Friedman RC, Farh KK, Burge CB and Bartel DP (2009) Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 19, 92-105 https://doi.org/10.1101/gr.082701.108
  29. Makarova JA, Shkurnikov MU, Wicklein D et al (2016) Intracellular and extracellular microRNA: an update on localization and biological role. Prog Histochem Cytochem 51, 33-49 https://doi.org/10.1016/j.proghi.2016.06.001
  30. Izarra A, Moscoso I, Levent E et al (2014) miR-133a enhances the protective capacity of cardiac progenitors cells after myocardial infarction. Stem Cell Rep 3, 1029-1042 https://doi.org/10.1016/j.stemcr.2014.10.010
  31. Zhou J, Li YS, Nguyen P et al (2013) Regulation of vascular smooth muscle cell turnover by endothelial cell-secreted microRNA-126: role of shear stress. Circ Res 113, 40-51 https://doi.org/10.1161/CIRCRESAHA.113.280883
  32. Rader DJ and Parmacek MS (2012) Secreted miRNAs suppress atherogenesis. Nat Cell Biol 14, 233-235 https://doi.org/10.1038/ncb2452
  33. Morel L, Regan M, Higashimori H et al (2013) Neuronal exosomal miRNA-dependent translational regulation of astroglial glutamate transporter GLT1. J Biol Chem 288, 7105-7116 https://doi.org/10.1074/jbc.M112.410944
  34. Ying W, Riopel M, Bandyopadhyay G et al (2017) Adipose tissue macrophage-derived exosomal miRNAs can modulate in vivo and in vitro insulin sensitivity. Cell 171, 372-384.e12 https://doi.org/10.1016/j.cell.2017.08.035
  35. Usman WM, Pham TC, Kwok YY et al (2018) Efficient RNA drug delivery using red blood cell extracellular vesicles. Nat Commun 9, 2359 https://doi.org/10.1038/s41467-018-04791-8
  36. Lou G, Song X, Yang F et al (2015) Exosomes derived from miR-122-modified adipose tissue-derived MSCs increase chemosensitivity of hepatocellular carcinoma. J Hematol Oncol 8, 122 https://doi.org/10.1186/s13045-015-0220-7
  37. Yu T, Zhao C, Hou S, Zhou W, Wang B and Chen Y (2019) Exosomes secreted from miRNA-29b-modified mesenchymal stem cells repaired spinal cord injury in rats. Braz J Med Biol Res 52, e8735 https://doi.org/10.1590/1414-431x20198735
  38. Lamichhane TN, Jeyaram A, Patel DB et al (2016) Oncogene knockdown via active loading of small RNAs into extracellular vesicles by sonication. Cell Mol Bioeng 9, 315-324 https://doi.org/10.1007/s12195-016-0457-4
  39. Liang G, Zhu Y, Ali DJ et al (2020) Engineered exosomes for targeted co-delivery of miR-21 inhibitor and chemotherapeutics to reverse drug resistance in colon cancer. J Nanobiotechnology 18, 10 https://doi.org/10.1186/s12951-019-0563-2
  40. Fu S, Wang Y, Xia X and Zheng JC (2020) Exosome engineering: current progress in cargo loading and targeted delivery. NanoImpact 20, 100261 https://doi.org/10.1016/j.impact.2020.100261
  41. Munir J, Yoon JK and Ryu S (2020) Therapeutic miRNA-enriched extracellular vesicles: current approaches and future prospects. Cells 9, 2271 https://doi.org/10.3390/cells9102271
  42. Zhang D, Lee H, Zhu Z, Minhas JK and Jin Y (2017) Enrichment of selective miRNAs in exosomes and delivery of exosomal miRNAs in vitro and in vivo. Am J Physiol Lung Cell Mol Physiol 312, L110-L121 https://doi.org/10.1152/ajplung.00423.2016
  43. Naseri Z, Oskuee RK, Forouzandeh-Moghadam M and Jaafari MR (2020) Delivery of LNA-antimiR-142-3p by mesenchymal stem cells-derived exosomes to breast cancer stem cells reduces tumorigenicity. Stem Cell Rev Rep 16, 541-556 https://doi.org/10.1007/s12015-019-09944-w
  44. Jeyaram A, Lamichhane TN, Wang S et al (2020) Enhanced loading of functional miRNA cargo via pH gradient modification of extracellular vesicles. Mol Ther 28, 975-985 https://doi.org/10.1016/j.ymthe.2019.12.007
  45. Peng Y, Zhao JL, Peng ZY, Xu WF and Yu GL (2020) Exosomal miR-25-3p from mesenchymal stem cells alleviates myocardial infarction by targeting pro-apoptotic proteins and EZH2. Cell Death Dis 11, 317 https://doi.org/10.1038/s41419-020-2545-6
  46. Ferguson SW, Wang J, Lee CJ et al (2018) The microRNA regulatory landscape of MSC-derived exosomes: a systems view. Sci Rep 8, 1419 https://doi.org/10.1038/s41598-018-19581-x
  47. Pan W, Xu X, Zhang M, Song X (2021) Human urine-derived stem cell-derived exosomal miR-21-5p promotes neurogenesis to attenuate Rett syndrome via the EPha4/TEK axis. Lab Invest 101, 824-836 https://doi.org/10.1038/s41374-021-00574-w
  48. Huang L, Fu C, Xiong F, He C and Wei Q (2021) Stem cell therapy for spinal cord injury. Cell Transplant 30, 963689721989266
  49. Hu J, Zeng L, Huang J, Wang G and Lu H (2015) miR-126 promotes angiogenesis and attenuates inflammation after contusion spinal cord injury in rats. Brain Res 1608, 191-202 https://doi.org/10.1016/j.brainres.2015.02.036
  50. Huang JH, Xu Y, Yin XM and Lin FY (2020) Exosomes derived from miR-126-modified MSCs promote angiogenesis and neurogenesis and attenuate apoptosis after spinal cord injury in rats. Neuroscience 424, 133-145 https://doi.org/10.1016/j.neuroscience.2019.10.043