Small Extracellular Vesicles Derived from Induced Pluripotent Stem Cells in the Treatment of Myocardial Injury
Abstract
1. Introduction
2. Isolation of sEVs from Induced Pluripotent Stem Cells
3. Drug Delivery of iPSC-sEVs in the Repair of Myocardial Injury
4. Mechanism of iPSC-sEVs in the Repair of Myocardial Injury
4.1. MI
4.2. MIRI
4.3. Coronary Heart Disease
4.4. HF
5. Challenges in the Treatment of CVD with IPSC-sEVs
6. Prospects and Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Kovacic, J.C.; Dimmeler, S.; Harvey, R.P.; Finkel, T.; Aikawa, E.; Krenning, G.; Baker, A.H. Endothelial to Mesenchymal Transition in Cardiovascular Disease: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2019, 73, 190–209. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Sureda, A.; Devkota, H.P.; Pittalà, V.; Barreca, D.; Silva, A.S.; Tewari, D.; Xu, S.; Nabavi, S.M. Curcumin, the golden spice in treating cardiovascular diseases. Biotechnol. Adv. 2020, 38, 107343. [Google Scholar] [CrossRef] [PubMed]
- Liu, C.; Du, L.; Wang, S.; Kong, L.; Zhang, S.; Li, S.; Zhang, W.; Du, G. Differences in the prevention and control of cardiovascular and cerebrovascular diseases. Pharmacol. Res. 2021, 170, 105737. [Google Scholar] [CrossRef]
- Pala, R.; Pattnaik, S.; Busi, S.; Nauli, S.M. Nanomaterials as Novel Cardiovascular Theranostics. Pharmaceutics 2021, 13, 348. [Google Scholar] [CrossRef]
- Terashvili, M.; Bosnjak, Z.J. Stem Cell Therapies in Cardiovascular Disease. J. Cardiothorac. Vasc. Anesthesia 2019, 33, 209–222. [Google Scholar] [CrossRef] [PubMed]
- Shi, Y.; Inoue, H.; Wu, J.C.; Yamanaka, S. Induced pluripotent stem cell technology: A decade of progress. Nat. Rev. Drug Discov. 2017, 16, 115–130. [Google Scholar] [CrossRef]
- Gao, P.; Liu, S.; Wang, X.; Ikeya, M. Dental applications of induced pluripotent stem cells and their derivatives. Jpn. Dent. Sci. Rev. 2022, 58, 162–171. [Google Scholar] [CrossRef]
- Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef]
- Deuse, T.; Hu, X.; Gravina, A.; Wang, D.; Tediashvili, G.; De, C.; Thayer, W.O.; Wahl, A.; Garcia, J.V.; Reichenspurner, H.; et al. Hypoimmunogenic derivatives of induced pluripotent stem cells evade immune rejection in fully immunocompetent allogeneic recipients. Nat. Biotechnol. 2019, 37, 252–258. [Google Scholar] [CrossRef]
- Doss, M.X.; Sachinidis, A. Current Challenges of iPSC-Based Disease Modeling and Therapeutic Implications. Cells 2019, 8, 403. [Google Scholar] [CrossRef]
- Crow, D. Could iPSCs Enable "Off-the-Shelf" Cell Therapy? Cell 2019, 177, 1667–1669. [Google Scholar] [CrossRef]
- Gao, L.; Wang, L.; Wei, Y.; Krishnamurthy, P.; Walcott, G.P.; Menasché, P.; Zhang, J. Exosomes secreted by hiPSC-derived cardiac cells improve recovery from myocardial infarction in swine. Sci. Transl. Med. 2020, 12, eaay1318. [Google Scholar] [CrossRef] [PubMed]
- Liu, L.; Michowski, W.; Kolodziejczyk, A.; Sicinski, P. The cell cycle in stem cell proliferation, pluripotency and differentiation. Nat. Cell Biol. 2019, 21, 1060–1067. [Google Scholar] [CrossRef] [PubMed]
- van der Pol, E.; Böing, A.N.; Harrison, P.; Sturk, A.; Nieuwland, R. Classification, functions, and clinical relevance of extracellular vesicles. Pharmacol. Rev. 2012, 64, 676–705. [Google Scholar] [CrossRef] [PubMed]
- Veerman, R.E.; Teeuwen, L.; Czarnewski, P.; Güclüler Akpinar, G.; Sandberg, A.; Cao, X.; Pernemalm, M.; Orre, L.M.; Gabrielsson, S.; Eldh, M. Molecular evaluation of five different isolation methods for extracellular vesicles reveals different clinical applicability and subcellular origin. J. Extracell. Vesicles 2021, 10, e12128. [Google Scholar] [CrossRef] [PubMed]
- Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef]
- Davidson, S.M.; Yellon, D.M. Exosomes and cardioprotection—A critical analysis. Mol. Asp. Med. 2018, 60, 104–114. [Google Scholar] [CrossRef]
- Maqsood, M.; Kang, M.; Wu, X.; Chen, J.; Teng, L.; Qiu, L. Adult mesenchymal stem cells and their exosomes: Sources, characteristics, and application in regenerative medicine. Life Sci. 2020, 256, 118002. [Google Scholar] [CrossRef]
- Yaghoubi, Y.; Movassaghpour, A.; Zamani, M.; Talebi, M.; Mehdizadeh, A.; Yousefi, M. Human umbilical cord mesenchymal stem cells derived-exosomes in diseases treatment. Life Sci. 2019, 233, 116733. [Google Scholar] [CrossRef]
- Peng, L.; Chen, Y.; Shi, S.; Wen, H. Stem cell-derived and circulating exosomal microRNAs as new potential tools for diabetic nephropathy management. Stem Cell Res. Ther. 2022, 13, 25. [Google Scholar] [CrossRef]
- Adamiak, M.; Cheng, G.; Bobis-Wozowicz, S.; Zhao, L.; Kedracka-Krok, S.; Samanta, A.; Karnas, E.; Xuan, Y.T.; Skupien-Rabian, B.; Chen, X.; et al. Induced Pluripotent Stem Cell (iPSC)-Derived Extracellular Vesicles Are Safer and More Effective for Cardiac Repair Than iPSCs. Circ. Res. 2018, 122, 296–309. [Google Scholar] [CrossRef] [PubMed]
- Goldenson, B.H.; Hor, P.; Kaufman, D.S. iPSC-Derived Natural Killer Cell Therapies—Expansion and Targeting. Front. Immunol. 2022, 13, 841107. [Google Scholar] [CrossRef] [PubMed]
- Mo, J.; Anastasaki, C.; Chen, Z.; Shipman, T.; Papke, J.; Yin, K.; Gutmann, D.H.; Le, L.Q. Humanized neurofibroma model from induced pluripotent stem cells delineates tumor pathogenesis and developmental origins. J. Clin. Investig. 2021, 131, e139807. [Google Scholar] [CrossRef]
- La Greca, A.; Solari, C.; Furmento, V.; Lombardi, A.; Biani, M.C.; Aban, C.; Moro, L.; García, M.; Guberman, A.S.; Sevlever, G.E.; et al. Extracellular vesicles from pluripotent stem cell-derived mesenchymal stem cells acquire a stromal modulatory proteomic pattern during differentiation. Exp. Mol. Med. 2018, 50, 1–12. [Google Scholar] [CrossRef]
- Hu, G.W.; Li, Q.; Niu, X.; Hu, B.; Liu, J.; Zhou, S.M.; Guo, S.C.; Lang, H.L.; Zhang, C.Q.; Wang, Y.; et al. Exosomes secreted by human-induced pluripotent stem cell-derived mesenchymal stem cells attenuate limb ischemia by promoting angiogenesis in mice. Stem Cell Res. Ther. 2015, 6, 10. [Google Scholar] [CrossRef] [PubMed]
- Sun, R.; Liu, Y.; Lu, M.; Ding, Q.; Wang, P.; Zhang, H.; Tian, X.; Lu, P.; Meng, D.; Sun, N.; et al. ALIX increases protein content and protective function of iPSC-derived exosomes. J. Mol. Med. 2019, 97, 829–844. [Google Scholar] [CrossRef]
- Kurtzwald-Josefson, E.; Zeevi-Levin, N.; Rubchevsky, V.; Bechar Erdman, N.; Schwartz Rohaker, O.; Nahum, O.; Hochhauser, E.; Ben-Avraham, B.; Itskovitz-Eldor, J.; Aravot, D.; et al. Cardiac Fibroblast-Induced Pluripotent Stem Cell-Derived Exosomes as a Potential Therapeutic Mean for Heart Failure. Int. J. Mol. Sci. 2020, 21, 7215. [Google Scholar] [CrossRef]
- Alzhrani, G.N.; Alanazi, S.T.; Alsharif, S.Y.; Albalawi, A.M.; Alsharif, A.A.; Abdel-Maksoud, M.S.; Elsherbiny, N. Exosomes: Isolation, characterization, and biomedical applications. Cell Biol. Int. 2021, 45, 1807–1831. [Google Scholar] [CrossRef]
- Gandham, S.; Su, X.; Wood, J.; Nocera, A.L.; Alli, S.C.; Milane, L.; Zimmerman, A.; Amiji, M.; Ivanov, A.R. Technologies and Standardization in Research on Extracellular Vesicles. Trends Biotechnol. 2020, 38, 1066–1098. [Google Scholar] [CrossRef] [PubMed]
- Chen, B.Y.; Sung, C.W.; Chen, C.; Cheng, C.M.; Lin, D.P.; Huang, C.T.; Hsu, M.Y. Advances in exosomes technology. Clin. Chim. Acta 2019, 493, 14–19. [Google Scholar] [CrossRef]
- Iwai, K.; Minamisawa, T.; Suga, K.; Yajima, Y.; Shiba, K. Isolation of human salivary extracellular vesicles by iodixanol density gradient ultracentrifugation and their characterizations. J. Extracell. Vesicles 2016, 5, 30829. [Google Scholar] [CrossRef] [PubMed]
- Böing, A.N.; van der Pol, E.; Grootemaat, A.E.; Coumans, F.A.; Sturk, A.; Nieuwland, R. Single-step isolation of extracellular vesicles by size-exclusion chromatography. J. Extracell. Vesicles 2014, 3, 23430. [Google Scholar] [CrossRef]
- Brown, P.N.; Yin, H. Polymer-Based Purification of Extracellular Vesicles. Methods Mol. Biol. 2017, 1660, 91–103. [Google Scholar] [CrossRef] [PubMed]
- Wijerathne, H.; Witek, M.A.; Jackson, J.M.; Brown, V.; Hupert, M.L.; Herrera, K.; Kramer, C.; Davidow, A.E.; Li, Y.; Baird, A.E.; et al. Affinity enrichment of extracellular vesicles from plasma reveals mRNA changes associated with acute ischemic stroke. Commun. Biol. 2020, 3, 613. [Google Scholar] [CrossRef]
- Wan, Y.; Cheng, G.; Liu, X.; Hao, S.J.; Nisic, M.; Zhu, C.D.; Xia, Y.Q.; Li, W.Q.; Wang, Z.G.; Zhang, W.L.; et al. Rapid magnetic isolation of extracellular vesicles via lipid-based nanoprobes. Nat. Biomed. Eng. 2017, 1, 0058. [Google Scholar] [CrossRef]
- Kim, Y.; Zhao, X. Magnetic Soft Materials and Robots. Chem. Rev. 2022, 122, 5317–5364. [Google Scholar] [CrossRef]
- Seo, N.; Nakamura, J.; Kaneda, T.; Tateno, H.; Shimoda, A.; Ichiki, T.; Furukawa, K.; Hirabayashi, J.; Akiyoshi, K.; Shiku, H. Distinguishing functional exosomes and other extracellular vesicles as a nucleic acid cargo by the anion-exchange method. J. Extracell. Vesicles 2022, 11, e12205. [Google Scholar] [CrossRef]
- Nordin, J.Z.; Lee, Y.; Vader, P.; Mäger, I.; Johansson, H.J.; Heusermann, W.; Wiklander, O.P.; Hällbrink, M.; Seow, Y.; Bultema, J.J.; et al. Ultrafiltration with size-exclusion liquid chromatography for high yield isolation of extracellular vesicles preserving intact biophysical and functional properties. Nanomedicine 2015, 11, 879–883. [Google Scholar] [CrossRef]
- Staubach, S.; Bauer, F.N.; Tertel, T.; Börger, V.; Stambouli, O.; Salzig, D.; Giebel, B. Scaled preparation of extracellular vesicles from conditioned media. Adv. Drug Deliv. Rev. 2021, 177, 113940. [Google Scholar] [CrossRef]
- Zhu, L.; Sun, H.T.; Wang, S.; Huang, S.L.; Zheng, Y.; Wang, C.Q.; Hu, B.Y.; Qin, W.; Zou, T.T.; Fu, Y.; et al. Isolation and characterization of exosomes for cancer research. J. Hematol. Oncol. 2020, 13, 152. [Google Scholar] [CrossRef]
- Zhu, L.; Kalimuthu, S.; Gangadaran, P.; Oh, J.M.; Lee, H.W.; Baek, S.H.; Jeong, S.Y.; Lee, S.W.; Lee, J.; Ahn, B.C. Exosomes Derived From Natural Killer Cells Exert Therapeutic Effect in Melanoma. Theranostics 2017, 7, 2732–2745. [Google Scholar] [CrossRef]
- Wang, J.; Ma, P.; Kim, D.H.; Liu, B.F.; Demirci, U. Towards Microfluidic-Based Exosome Isolation and Detection for Tumor Therapy. Nano Today 2021, 37, 101066. [Google Scholar] [CrossRef] [PubMed]
- Bo, Y.; Yang, L.; Liu, B.; Tian, G.; Li, C.; Zhang, L.; Yan, Y. Exosomes from human induced pluripotent stem cells-derived keratinocytes accelerate burn wound healing through miR-762 mediated promotion of keratinocytes and endothelial cells migration. J. Nanobiotechnol. 2022, 20, 291. [Google Scholar] [CrossRef] [PubMed]
- Marzano, M.; Bejoy, J.; Cheerathodi, M.R.; Sun, L.; York, S.B.; Zhao, J.; Kanekiyo, T.; Bu, G.; Meckes, D.G., Jr.; Li, Y. Differential Effects of Extracellular Vesicles of Lineage-Specific Human Pluripotent Stem Cells on the Cellular Behaviors of Isogenic Cortical Spheroids. Cells 2019, 8, 993. [Google Scholar] [CrossRef]
- Gardiner, C.; Ferreira, Y.J.; Dragovic, R.A.; Redman, C.W.; Sargent, I.L. Extracellular vesicle sizing and enumeration by nanoparticle tracking analysis. J. Extracell. Vesicles 2013, 2, 19671. [Google Scholar] [CrossRef] [PubMed]
- Lobb, R.J.; Becker, M.; Wen, S.W.; Wong, C.S.; Wiegmans, A.P.; Leimgruber, A.; Möller, A. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J. Extracell. Vesicles 2015, 4, 27031. [Google Scholar] [CrossRef] [PubMed]
- Ueda, K.; Ishikawa, N.; Tatsuguchi, A.; Saichi, N.; Fujii, R.; Nakagawa, H. Antibody-coupled monolithic silica microtips for highthroughput molecular profiling of circulating exosomes. Sci. Rep. 2014, 4, 6232. [Google Scholar] [CrossRef]
- Pocsfalvi, G.; Stanly, C.; Vilasi, A.; Fiume, I.; Capasso, G.; Turiák, L.; Buzas, E.I.; Vékey, K. Mass spectrometry of extracellular vesicles. Mass Spectrom. Rev. 2016, 35, 3–21. [Google Scholar] [CrossRef]
- de Rus Jacquet, A.; Tancredi, J.L.; Lemire, A.L.; DeSantis, M.C.; Li, W.P.; O’Shea, E.K. The LRRK2 G2019S mutation alters astrocyte-to-neuron communication via extracellular vesicles and induces neuron atrophy in a human iPSC-derived model of Parkinson’s disease. eLife 2021, 10, e73062. [Google Scholar] [CrossRef]
- Street, J.M.; Barran, P.E.; Mackay, C.L.; Weidt, S.; Balmforth, C.; Walsh, T.S.; Chalmers, R.T.; Webb, D.J.; Dear, J.W. Identification and proteomic profiling of exosomes in human cerebrospinal fluid. J. Transl. Med. 2012, 10, 5. [Google Scholar] [CrossRef]
- Chen, J.; Li, P.; Zhang, T.; Xu, Z.; Huang, X.; Wang, R.; Du, L. Review on Strategies and Technologies for Exosome Isolation and Purification. Front. Bioeng. Biotechnol. 2021, 9, 811971. [Google Scholar] [CrossRef] [PubMed]
- Dong, L.; Zieren, R.C.; Horie, K.; Kim, C.J.; Mallick, E.; Jing, Y.; Feng, M.; Kuczler, M.D.; Green, J.; Amend, S.R.; et al. Comprehensive evaluation of methods for small extracellular vesicles separation from human plasma, urine and cell culture medium. J. Extracell. Vesicles 2020, 10, e12044. [Google Scholar] [CrossRef]
- Koh, Y.Q.; Almughlliq, F.B.; Vaswani, K.; Peiris, H.N.; Mitchell, M.D. Exosome enrichment by ultracentrifugation and size exclusion chromatography. Front. Biosci. (Landmark Ed.) 2018, 23, 865–874. [Google Scholar] [CrossRef]
- Li, S.; Yi, M.; Dong, B.; Tan, X.; Luo, S.; Wu, K. The role of exosomes in liquid biopsy for cancer diagnosis and prognosis prediction. Int. J. Cancer 2021, 148, 2640–2651. [Google Scholar] [CrossRef]
- Yu, D.; Li, Y.; Wang, M.; Gu, J.; Xu, W.; Cai, H.; Fang, X.; Zhang, X. Exosomes as a new frontier of cancer liquid biopsy. Mol. Cancer 2022, 21, 56. [Google Scholar] [CrossRef]
- Northrop-Albrecht, E.J.; Taylor, W.R.; Huang, B.Q.; Kisiel, J.B.; Lucien, F. Assessment of extracellular vesicle isolation methods from human stool supernatant. J. Extracell. Vesicles 2022, 11, e12208. [Google Scholar] [CrossRef]
- Onódi, Z.; Pelyhe, C.; Terézia Nagy, C.; Brenner, G.B.; Almási, L.; Kittel, Á.; Manček-Keber, M.; Ferdinandy, P.; Buzás, E.I.; Giricz, Z. Isolation of High-Purity Extracellular Vesicles by the Combination of Iodixanol Density Gradient Ultracentrifugation and Bind-Elute Chromatography From Blood Plasma. Front. Physiol. 2018, 9, 1479. [Google Scholar] [CrossRef]
- Liu, Y.; Xu, C.; Gao, T.; Chen, X.; Wang, J.; Yu, P.; Mao, L. Sizing Single Particles at the Orifice of a Nanopipette. ACS Sens. 2020, 5, 2351–2358. [Google Scholar] [CrossRef]
- Guo, J.; Wu, C.; Lin, X.; Zhou, J.; Zhang, J.; Zheng, W.; Wang, T.; Cui, Y. Establishment of a simplified dichotomic size-exclusion chromatography for isolating extracellular vesicles toward clinical applications. J. Extracell. Vesicles 2021, 10, e12145. [Google Scholar] [CrossRef]
- Karimi, N.; Cvjetkovic, A.; Jang, S.C.; Crescitelli, R.; Hosseinpour Feizi, M.A.; Nieuwland, R.; Lötvall, J.; Lässer, C. Detailed analysis of the plasma extracellular vesicle proteome after separation from lipoproteins. Cell. Mol. Life Sci. 2018, 75, 2873–2886. [Google Scholar] [CrossRef]
- Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef] [PubMed]
- Fitzner, D.; Schnaars, M.; van Rossum, D.; Krishnamoorthy, G.; Dibaj, P.; Bakhti, M.; Regen, T.; Hanisch, U.K.; Simons, M. Selective transfer of exosomes from oligodendrocytes to microglia by macropinocytosis. J. Cell Sci. 2011, 124, 447–458. [Google Scholar] [CrossRef] [PubMed]
- Tkach, M.; Théry, C. Communication by Extracellular Vesicles: Where We Are and Where We Need to Go. Cell 2016, 164, 1226–1232. [Google Scholar] [CrossRef]
- Ostrowski, M.; Carmo, N.B.; Krumeich, S.; Fanget, I.; Raposo, G.; Savina, A.; Moita, C.F.; Schauer, K.; Hume, A.N.; Freitas, R.P.; et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat. Cell Biol. 2010, 12, 19–30. [Google Scholar] [CrossRef]
- Hsu, C.; Morohashi, Y.; Yoshimura, S.; Manrique-Hoyos, N.; Jung, S.; Lauterbach, M.A.; Bakhti, M.; Grønborg, M.; Möbius, W.; Rhee, J.; et al. Regulation of exosome secretion by Rab35 and its GTPase-activating proteins TBC1D10A-C. J. Cell Biol. 2010, 189, 223–232. [Google Scholar] [CrossRef] [PubMed]
- Chutna, O.; Gonçalves, S.; Villar-Piqué, A.; Guerreiro, P.; Marijanovic, Z.; Mendes, T.; Ramalho, J.; Emmanouilidou, E.; Ventura, S.; Klucken, J.; et al. The small GTPase Rab11 co-localizes with α-synuclein in intracellular inclusions and modulates its aggregation, secretion and toxicity. Hum. Mol. Genet. 2014, 23, 6732–6745. [Google Scholar] [CrossRef]
- Sung, B.H.; Ketova, T.; Hoshino, D.; Zijlstra, A.; Weaver, A.M. Directional cell movement through tissues is controlled by exosome secretion. Nat. Commun. 2015, 6, 7164. [Google Scholar] [CrossRef]
- Bobrie, A.; Krumeich, S.; Reyal, F.; Recchi, C.; Moita, L.F.; Seabra, M.C.; Ostrowski, M.; Théry, C. Rab27a supports exosome-dependent and -independent mechanisms that modify the tumor microenvironment and can promote tumor progression. Cancer Res. 2012, 72, 4920–4930. [Google Scholar] [CrossRef]
- Lai, C.P.; Mardini, O.; Ericsson, M.; Prabhakar, S.; Maguire, C.; Chen, J.W.; Tannous, B.A.; Breakefield, X.O. Dynamic biodistribution of extracellular vesicles in vivo using a multimodal imaging reporter. ACS Nano 2014, 8, 483–494. [Google Scholar] [CrossRef]
- Lai, C.P.; Kim, E.Y.; Badr, C.E.; Weissleder, R.; Mempel, T.R.; Tannous, B.A.; Breakefield, X.O. Visualization and tracking of tumour extracellular vesicle delivery and RNA translation using multiplexed reporters. Nat. Commun. 2015, 6, 7029. [Google Scholar] [CrossRef] [PubMed]
- Lu, M.; Peng, L.; Ming, X.; Wang, X.; Cui, A.; Li, Y.; Wang, X.; Meng, D.; Sun, N.; Xiang, M.; et al. Enhanced wound healing promotion by immune response-free monkey autologous iPSCs and exosomes vs. their allogeneic counterparts. EBioMedicine 2019, 42, 443–457. [Google Scholar] [CrossRef]
- Pegtel, D.M.; Cosmopoulos, K.; Thorley-Lawson, D.A.; van Eijndhoven, M.A.; Hopmans, E.S.; Lindenberg, J.L.; de Gruijl, T.D.; Würdinger, T.; Middeldorp, J.M. Functional delivery of viral miRNAs via exosomes. Proc. Natl. Acad. Sci. USA 2010, 107, 6328–6333. [Google Scholar] [CrossRef]
- Mendell, J.T. miRiad roles for the miR-17-92 cluster in development and disease. Cell 2008, 133, 217–222. [Google Scholar] [CrossRef]
- Armstrong, J.P.; Holme, M.N.; Stevens, M.M. Re-Engineering Extracellular Vesicles as Smart Nanoscale Therapeutics. ACS Nano 2017, 11, 69–83. [Google Scholar] [CrossRef]
- Luo, R.; Liu, M.; Tan, T.; Yang, Q.; Wang, Y.; Men, L.; Zhao, L.; Zhang, H.; Wang, S.; Xie, T.; et al. Emerging Significance and Therapeutic Potential of Extracellular vesicles. Int. J. Biol. Sci. 2021, 17, 2476–2486. [Google Scholar] [CrossRef]
- Xitong, D.; Xiaorong, Z. Targeted therapeutic delivery using engineered exosomes and its applications in cardiovascular diseases. Gene 2016, 575, 377–384. [Google Scholar] [CrossRef]
- Bobis-Wozowicz, S.; Kmiotek, K.; Sekula, M.; Kedracka-Krok, S.; Kamycka, E.; Adamiak, M.; Jankowska, U.; Madetko-Talowska, A.; Sarna, M.; Bik-Multanowski, M.; et al. Human Induced Pluripotent Stem Cell-Derived Microvesicles Transmit RNAs and Proteins to Recipient Mature Heart Cells Modulating Cell Fate and Behavior. Stem Cells 2015, 33, 2748–2761. [Google Scholar] [CrossRef]
- Zhang, L.L.; Liu, J.J.; Liu, F.; Liu, W.H.; Wang, Y.S.; Zhu, B.; Yu, B. MiR-499 induces cardiac differentiation of rat mesenchymal stem cells through wnt/β-catenin signaling pathway. Biochem. Biophys. Res. Commun. 2012, 420, 875–881. [Google Scholar] [CrossRef]
- Jung, J.H.; Ikeda, G.; Tada, Y.; von Bornstädt, D.; Santoso, M.R.; Wahlquist, C.; Rhee, S.; Jeon, Y.J.; Yu, A.C.; O’Brien, C.G.; et al. miR-106a-363 cluster in extracellular vesicles promotes endogenous myocardial repair via Notch3 pathway in ischemic heart injury. Basic Res. Cardiol. 2021, 116, 19. [Google Scholar] [CrossRef]
- Luo, Y.; Gao, D.; Wang, P.; Lou, C.; Li, T.; Niu, W.; Gao, Y. Optimized culture methods for isolating small extracellular vesicles derived from human induced pluripotent stem cells. J. Extracell. Vesicles 2021, 10, e12065. [Google Scholar] [CrossRef]
- Zhu, D.; Li, Z.; Huang, K.; Caranasos, T.G.; Rossi, J.S.; Cheng, K. Minimally invasive delivery of therapeutic agents by hydrogel injection into the pericardial cavity for cardiac repair. Nat. Commun. 2021, 12, 1412. [Google Scholar] [CrossRef]
- Wang, Y.; Zhang, L.; Li, Y.; Chen, L.; Wang, X.; Guo, W.; Zhang, X.; Qin, G.; He, S.H.; Zimmerman, A.; et al. Exosomes/microvesicles from induced pluripotent stem cells deliver cardioprotective miRNAs and prevent cardiomyocyte apoptosis in the ischemic myocardium. Int. J. Cardiol. 2015, 192, 61–69. [Google Scholar] [CrossRef]
- Takeda, M.; Kanki, Y.; Masumoto, H.; Funakoshi, S.; Hatani, T.; Fukushima, H.; Izumi-Taguchi, A.; Matsui, Y.; Shimamura, T.; Yoshida, Y.; et al. Identification of Cardiomyocyte-Fated Progenitors from Human-Induced Pluripotent Stem Cells Marked with CD82. Cell Rep. 2018, 22, 546–556. [Google Scholar] [CrossRef]
- Ellis, B.W.; Ronan, G.; Ren, X.; Bahcecioglu, G.; Senapati, S.; Anderson, D.; Handberg, E.; March, K.L.; Chang, H.C.; Zorlutuna, P. Human Heart Anoxia and Reperfusion Tissue (HEART) Model for the Rapid Study of Exosome Bound miRNA Expression As Biomarkers for Myocardial Infarction. Small 2022, 18, e2201330. [Google Scholar] [CrossRef]
- Jiang, X.; Sucharov, J.; Stauffer, B.L.; Miyamoto, S.D.; Sucharov, C.C. Exosomes from pediatric dilated cardiomyopathy patients modulate a pathological response in cardiomyocytes. Am. J. Physiol. Circ. Physiol. 2017, 312, H818–H826. [Google Scholar] [CrossRef]
- Turner, A.; Aggarwal, P.; Matter, A.; Olson, B.; Gu, C.C.; Hunt, S.C.; Lewis, C.E.; Arnett, D.K.; Lorier, R.; Broeckel, U. Donor-specific phenotypic variation in hiPSC cardiomyocyte-derived exosomes impacts endothelial cell function. Am. J. Physiol. Circ. Physiol. 2021, 320, H954–H968. [Google Scholar] [CrossRef]
- Srivastava, D.; Ivey, K.N. Potential of stem-cell-based therapies for heart disease. Nature 2006, 441, 1097–1099. [Google Scholar] [CrossRef]
- Gartz, M.; Darlington, A.; Afzal, M.Z.; Strande, J.L. Exosomes exert cardioprotection in dystrophin-deficient cardiomyocytes via ERK1/2-p38/MAPK signaling. Sci. Rep. 2018, 8, 16519. [Google Scholar] [CrossRef]
- Lee, W.H.; Chen, W.Y.; Shao, N.Y.; Xiao, D.; Qin, X.; Baker, N.; Bae, H.R.; Wei, T.T.; Wang, Y.; Shukla, P.; et al. Comparison of Non-Coding RNAs in Exosomes and Functional Efficacy of Human Embryonic Stem Cell- versus Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Stem Cells 2017, 35, 2138–2149. [Google Scholar] [CrossRef]
- Chen, M.; Chen, J.; Huang, W.; Li, C.; Luo, H.; Xue, Z.; Xiao, Y.; Wu, Q.; Chen, C. Exosomes from human induced pluripotent stem cells derived mesenchymal stem cells improved myocardial injury caused by severe acute pancreatitis through activating Akt/Nrf2/HO-1 axis. Cell Cycle 2022, 21, 1578–1589. [Google Scholar] [CrossRef]
- Zhang, J.; Guan, J.; Niu, X.; Hu, G.; Guo, S.; Li, Q.; Xie, Z.; Zhang, C.; Wang, Y. Exosomes released from human induced pluripotent stem cells-derived MSCs facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis. J. Transl. Med. 2015, 13, 49. [Google Scholar] [CrossRef]
- El Harane, N.; Kervadec, A.; Bellamy, V.; Pidial, L.; Neametalla, H.J.; Perier, M.-C.; Lima Correa, B.; Thiébault, L.; Cagnard, N.; Duché, A.; et al. Acellular therapeutic approach for heart failure: In vitro production of extracellular vesicles from human cardiovascular progenitors. Eur. Heart J. 2018, 39, 1835–1847. [Google Scholar] [CrossRef] [PubMed]
- Mayourian, J.; Ceholski, D.K.; Gorski, P.A.; Mathiyalagan, P.; Murphy, J.F.; Salazar, S.I.; Stillitano, F.; Hare, J.M.; Sahoo, S.; Hajjar, R.J.; et al. Exosomal microRNA-21-5p Mediates Mesenchymal Stem Cell Paracrine Effects on Human Cardiac Tissue Contractility. Circ. Res. 2018, 122, 933–944. [Google Scholar] [CrossRef]
- Jung, J.H.; Fu, X.; Yang, P.C. Exosomes Generated From iPSC-Derivatives: New Direction for Stem Cell Therapy in Human Heart Diseases. Circ. Res. 2017, 120, 407–417. [Google Scholar] [CrossRef]
- Yuan, X.; Li, D.; Chen, X.; Han, C.; Xu, L.; Huang, T.; Dong, Z.; Zhang, M. Extracellular vesicles from human-induced pluripotent stem cell-derived mesenchymal stromal cells (hiPSC-MSCs) protect against renal ischemia/reperfusion injury via delivering specificity protein (SP1) and transcriptional activating of sphingosine kinase 1 and inhibiting necroptosis. Cell Death Dis. 2017, 8, 3200. [Google Scholar] [CrossRef]
- Dougherty, J.A.; Kumar, N.; Noor, M.; Angelos, M.G.; Khan, M.; Chen, C.A.; Khan, M. Extracellular Vesicles Released by Human Induced-Pluripotent Stem Cell-Derived Cardiomyocytes Promote Angiogenesis. Front. Physiol. 2018, 9, 1794. [Google Scholar] [CrossRef]
- Zununi Vahed, S.; Barzegari, A.; Zuluaga, M.; Letourneur, D.; Pavon-Djavid, G. Myocardial infarction and gut microbiota: An incidental connection. Pharmacol. Res. 2018, 129, 308–317. [Google Scholar] [CrossRef]
- Zhao, M.; Nakada, Y.; Wei, Y.; Bian, W.; Chu, Y.; Borovjagin, A.V.; Xie, M.; Zhu, W.; Nguyen, T.; Zhou, Y.; et al. Cyclin D2 Overexpression Enhances the Efficacy of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Myocardial Repair in a Swine Model of Myocardial Infarction. Circulation 2021, 144, 210–228. [Google Scholar] [CrossRef]
- Lozano, J.; Rai, A.; Lees, J.G.; Fang, H.; Claridge, B.; Lim, S.Y.; Greening, D.W. Scalable Generation of Nanovesicles from Human-Induced Pluripotent Stem Cells for Cardiac Repair. Int. J. Mol. Sci. 2022, 23, 14334. [Google Scholar] [CrossRef]
- Sharma, A.; Zhang, Y.; Buikema, J.W.; Serpooshan, V.; Chirikian, O.; Kosaric, N.; Churko, J.M.; Dzilic, E.; Shieh, A.; Burridge, P.W.; et al. Stage-specific Effects of Bioactive Lipids on Human iPSC Cardiac Differentiation and Cardiomyocyte Proliferation. Sci. Rep. 2018, 8, 6618. [Google Scholar] [CrossRef]
- Gao, L.; Gregorich, Z.R.; Zhu, W.; Mattapally, S.; Oduk, Y.; Lou, X.; Kannappan, R.; Borovjagin, A.V.; Walcott, G.P.; Pollard, A.E.; et al. Large Cardiac Muscle Patches Engineered From Human Induced-Pluripotent Stem Cell-Derived Cardiac Cells Improve Recovery From Myocardial Infarction in Swine. Circulation 2018, 137, 1712–1730. [Google Scholar] [CrossRef] [PubMed]
- Ibrahim, A.G.; Cheng, K.; Marbán, E. Exosomes as critical agents of cardiac regeneration triggered by cell therapy. Stem Cell Rep. 2014, 2, 606–619. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Ha, T.; Liu, L.; Zou, J.; Zhang, X.; Kalbfleisch, J.; Gao, X.; Williams, D.; Li, C. Increased expression of microRNA-146a decreases myocardial ischaemia/reperfusion injury. Cardiovasc. Res. 2013, 97, 432–442. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Liew, O.W.; Richards, A.M.; Chen, Y.T. Overview of MicroRNAs in Cardiac Hypertrophy, Fibrosis, and Apoptosis. Int. J. Mol. Sci. 2016, 17, 749. [Google Scholar] [CrossRef] [PubMed]
- Zhu, H.; Fan, G.C. Extracellular/circulating microRNAs and their potential role in cardiovascular disease. Am. J. Cardiovasc. Dis. 2011, 1, 138–149. [Google Scholar] [PubMed]
- Huang, W.; Tian, S.S.; Hang, P.Z.; Sun, C.; Guo, J.; Du, Z.M. Combination of microRNA-21 and microRNA-146a Attenuates Cardiac Dysfunction and Apoptosis During Acute Myocardial Infarction in Mice. Mol. Ther. Nucleic Acids 2016, 5, e296. [Google Scholar] [CrossRef]
- Fang, Y.H.; Wang, S.P.H.; Gao, Z.H.; Wu, S.N.; Chang, H.Y.; Yang, P.J.; Liu, P.Y.; Liu, Y.W. Efficient Cardiac Differentiation of Human Amniotic Fluid-Derived Stem Cells into Induced Pluripotent Stem Cells and Their Potential Immune Privilege. Int. J. Mol. Sci. 2020, 21, 2359. [Google Scholar] [CrossRef]
- Taheri, B.; Soleimani, M.; Fekri Aval, S.; Esmaeili, E.; Bazi, Z.; Zarghami, N. Induced pluripotent stem cell-derived extracellular vesicles: A novel approach for cell-free regenerative medicine. J. Cell. Physiol. 2019, 234, 8455–8464. [Google Scholar] [CrossRef]
- Deanfield, J.E.; Halcox, J.P.; Rabelink, T.J. Endothelial function and dysfunction: Testing and clinical relevance. Circulation 2007, 115, 1285–1295. [Google Scholar] [CrossRef]
- Sun, X.; Feinberg, M.W. Regulation of endothelial cell metabolism: Just go with the flow. Arter. Thromb. Vasc. Biol. 2015, 35, 13–15. [Google Scholar] [CrossRef]
- Jouda, H.; Larrea Murillo, L.; Wang, T. Current Progress in Vascular Engineering and Its Clinical Applications. Cells 2022, 11, 493. [Google Scholar] [CrossRef] [PubMed]
- van Rooij, E.; Olson, E.N. MicroRNA therapeutics for cardiovascular disease: Opportunities and obstacles. Nat. Rev. Drug Discov. 2012, 11, 860–872. [Google Scholar] [CrossRef] [PubMed]
- de Couto, G.; Gallet, R.; Cambier, L.; Jaghatspanyan, E.; Makkar, N.; Dawkins, J.F.; Berman, B.P.; Marbán, E. Exosomal MicroRNA Transfer Into Macrophages Mediates Cellular Postconditioning. Circulation 2017, 136, 200–214. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Han, Z.; Yu, Y.; Xu, Z.; Cai, B.; Yuan, Y. Potential Applications of Induced Pluripotent Stem Cells for Cardiovascular Diseases. Curr. Drug Targets 2019, 20, 763–774. [Google Scholar] [CrossRef]
- de Godoy, M.A.; Saraiva, L.M.; de Carvalho, L.R.P.; Vasconcelos-Dos-Santos, A.; Beiral, H.J.V.; Ramos, A.B.; Silva, L.R.P.; Leal, R.B.; Monteiro, V.H.S.; Braga, C.V.; et al. Mesenchymal stem cells and cell-derived extracellular vesicles protect hippocampal neurons from oxidative stress and synapse damage induced by amyloid-β oligomers. J. Biol. Chem. 2018, 293, 1957–1975. [Google Scholar] [CrossRef] [PubMed]
- Ding, Q.; Sun, R.; Wang, P.; Zhang, H.; Xiang, M.; Meng, D.; Sun, N.; Chen, A.F.; Chen, S. Protective effects of human induced pluripotent stem cell-derived exosomes on high glucose-induced injury in human endothelial cells. Exp. Ther. Med. 2018, 15, 4791–4797. [Google Scholar] [CrossRef]
- Gupta, S.; Krishnakumar, V.; Soni, N.; Rao, E.P.; Banerjee, A.; Mohanty, S. Comparative proteomic profiling of Small Extracellular vesicles derived from iPSCs and tissue specific mesenchymal stem cells. Exp. Cell Res. 2022, 420, 113354. [Google Scholar] [CrossRef]
- Barreca, M.M.; Cancemi, P.; Geraci, F. Mesenchymal and Induced Pluripotent Stem Cells-Derived Extracellular Vesicles: The New Frontier for Regenerative Medicine? Cells 2020, 9, 1163. [Google Scholar] [CrossRef]
- Hunt, S.A. Current status of cardiac transplantation. JAMA 1998, 280, 1692–1698. [Google Scholar] [CrossRef]
- Tian, C.; Gao, L.; Zucker, I.H. Regulation of Nrf2 signaling pathway in heart failure: Role of extracellular vesicles and non-coding RNAs. Free. Radic. Biol. Med. 2021, 167, 218–231. [Google Scholar] [CrossRef]
- Qiao, L.; Hu, S.; Liu, S.; Zhang, H.; Ma, H.; Huang, K.; Li, Z.; Su, T.; Vandergriff, A.; Tang, J.; et al. microRNA-21-5p dysregulation in exosomes derived from heart failure patients impairs regenerative potential. J. Clin. Investig. 2019, 129, 2237–2250. [Google Scholar] [CrossRef] [PubMed]
- Bär, C.; Chatterjee, S.; Falcão Pires, I.; Rodrigues, P.; Sluijter, J.P.G.; Boon, R.A.; Nevado, R.M.; Andrés, V.; Sansonetti, M.; de Windt, L.; et al. Non-coding RNAs: Update on mechanisms and therapeutic targets from the ESC Working Groups of Myocardial Function and Cellular Biology of the Heart. Cardiovasc. Res. 2020, 116, 1805–1819. [Google Scholar] [CrossRef] [PubMed]
- Viereck, J.; Bührke, A.; Foinquinos, A.; Chatterjee, S.; Kleeberger, J.A.; Xiao, K.; Janssen-Peters, H.; Batkai, S.; Ramanujam, D.; Kraft, T.; et al. Targeting muscle-enriched long non-coding RNA H19 reverses pathological cardiac hypertrophy. Eur. Heart J. 2020, 41, 3462–3474. [Google Scholar] [CrossRef] [PubMed]
- Hou, J.R.; Wang, Y.H.; Zhong, Y.N.; Che, T.T.; Hu, Y.; Bao, J.; Meng, N. Protective Effect of Flavonoids from a Deep-Sea-Derived Arthrinium sp. against ox-LDL-Induced Oxidative Injury through Activating the AKT/Nrf2/HO-1 Pathway in Vascular Endothelial Cells. Mar. Drugs 2021, 19, 712. [Google Scholar] [CrossRef] [PubMed]
- Karnas, E.; Sekuła-Stryjewska, M.; Kmiotek-Wasylewska, K.; Bobis-Wozowicz, S.; Ryszawy, D.; Sarna, M.; Madeja, Z.; Zuba-Surma, E.K. Extracellular vesicles from human iPSCs enhance reconstitution capacity of cord blood-derived hematopoietic stem and progenitor cells. Leukemia 2021, 35, 2964–2977. [Google Scholar] [CrossRef]
- Upadhya, R.; Madhu, L.N.; Rao, S.; Shetty, A.K. Proficiency of Extracellular Vesicles From hiPSC-Derived Neural Stem Cells in Modulating Proinflammatory Human Microglia: Role of Pentraxin-3 and miRNA-21-5p. Front. Mol. Neurosci. 2022, 15, 845542. [Google Scholar] [CrossRef]
- Chandy, M.; Rhee, J.W.; Ozen, M.O.; Williams, D.R.; Pepic, L.; Liu, C.; Zhang, H.; Malisa, J.; Lau, E.; Demirci, U.; et al. Atlas of Exosomal microRNAs Secreted From Human iPSC-Derived Cardiac Cell Types. Circulation 2020, 142, 1794–1796. [Google Scholar] [CrossRef]
- Goversen, B.; van der Heyden, M.A.G.; van Veen, T.A.B.; de Boer, T.P. The immature electrophysiological phenotype of iPSC-CMs still hampers in vitro drug screening: Special focus on I(K1). Pharmacol. Ther. 2018, 183, 127–136. [Google Scholar] [CrossRef]
- Ronaldson-Bouchard, K.; Ma, S.P.; Yeager, K.; Chen, T.; Song, L.; Sirabella, D.; Morikawa, K.; Teles, D.; Yazawa, M.; Vunjak-Novakovic, G. Advanced maturation of human cardiac tissue grown from pluripotent stem cells. Nature 2018, 556, 239–243. [Google Scholar] [CrossRef]
- Karbassi, E.; Fenix, A.; Marchiano, S.; Muraoka, N.; Nakamura, K.; Yang, X.; Murry, C.E. Cardiomyocyte maturation: Advances in knowledge and implications for regenerative medicine. Nat. Rev. Cardiol. 2020, 17, 341–359. [Google Scholar] [CrossRef]
- Chow, M.; Boheler, K.R.; Li, R.A. Human pluripotent stem cell-derived cardiomyocytes for heart regeneration, drug discovery and disease modeling: From the genetic, epigenetic, and tissue modeling perspectives. Stem Cell Res. Ther. 2013, 4, 97. [Google Scholar] [CrossRef]
- Hollý, D.; Klein, M.; Mazreku, M.; Zamborský, R.; Polák, Š.; Danišovič, Ľ.; Csöbönyeiová, M. Stem Cells and Their Derivatives-Implications for Alveolar Bone Regeneration: A Comprehensive Review. Int. J. Mol. Sci. 2021, 22, 11746. [Google Scholar] [CrossRef] [PubMed]
- Yu, L.; Yang, Y.; Zhang, B.; Bai, X.; Fei, Q.; Zhang, L. Rapid human-derived iPSC osteogenesis combined with three-dimensionally printed Ti6Al4V scaffolds for the repair of bone defects. J. Cell. Physiol. 2020, 235, 9763–9772. [Google Scholar] [CrossRef] [PubMed]
- Bec, N.; Bonhoure, A.; Henry, L.; Berry, L.; Larroque, C.; Coux, O.; Stoebner, P.E.; Vidal, M. Proteasome 19S RP and translation preinitiation complexes are secreted within exosomes upon serum starvation. Traffic 2019, 20, 516–536. [Google Scholar] [CrossRef]
- Bister, N.; Pistono, C.; Huremagic, B.; Jolkkonen, J.; Giugno, R.; Malm, T. Hypoxia and extracellular vesicles: A review on methods, vesicular cargo and functions. J. Extracell. Vesicles 2020, 10, e12002. [Google Scholar] [CrossRef] [PubMed]
- Nachlas, A.L.Y.; Li, S.; Jha, R.; Singh, M.; Xu, C.; Davis, M.E. Human iPSC-derived mesenchymal stem cells encapsulated in PEGDA hydrogels mature into valve interstitial-like cells. Acta Biomater. 2018, 71, 235–246. [Google Scholar] [CrossRef]
- Noor, N.; Shapira, A.; Edri, R.; Gal, I.; Wertheim, L.; Dvir, T. 3D Printing of Personalized Thick and Perfusable Cardiac Patches and Hearts. Adv. Sci. 2019, 6, 1900344. [Google Scholar] [CrossRef] [PubMed]
- Tabei, R.; Kawaguchi, S.; Kanazawa, H.; Tohyama, S.; Hirano, A.; Handa, N.; Hishikawa, S.; Teratani, T.; Kunita, S.; Fukuda, J.; et al. Development of a transplant injection device for optimal distribution and retention of human induced pluripotent stem cell–derived cardiomyocytes. J. Heart Lung Transplant. 2019, 38, 203–214. [Google Scholar] [CrossRef]
- Qi, X.; Zhang, J.; Yuan, H.; Xu, Z.; Li, Q.; Niu, X.; Hu, B.; Wang, Y.; Li, X. Exosomes Secreted by Human-Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cells Repair Critical-Sized Bone Defects through Enhanced Angiogenesis and Osteogenesis in Osteoporotic Rats. Int. J. Biol. Sci. 2016, 12, 836–849. [Google Scholar] [CrossRef]
- Tian, T.; Wang, Y.; Wang, H.; Zhu, Z.; Xiao, Z. Visualizing of the cellular uptake and intracellular trafficking of exosomes by live-cell microscopy. J. Cell. Biochem. 2010, 111, 488–496. [Google Scholar] [CrossRef]
- Feng, D.; Zhao, W.L.; Ye, Y.Y.; Bai, X.C.; Liu, R.Q.; Chang, L.F.; Zhou, Q.; Sui, S.F. Cellular internalization of exosomes occurs through phagocytosis. Traffic 2010, 11, 675–687. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.; Xue, R.; Huang, P.; Wu, Y.; Fan, W.; He, X.; Dong, Y.; Liu, C. Modified Exosomes: A Good Transporter for miRNAs within Stem Cells to Treat Ischemic Heart Disease. J. Cardiovasc. Transl. Res. 2022, 15, 514–523. [Google Scholar] [CrossRef] [PubMed]
- Holm, F.; Ström, S.; Inzunza, J.; Baker, D.; Strömberg, A.M.; Rozell, B.; Feki, A.; Bergström, R.; Hovatta, O. An effective serum- and xeno-free chemically defined freezing procedure for human embryonic and induced pluripotent stem cells. Hum. Reprod. 2010, 25, 1271–1279. [Google Scholar] [CrossRef]
- Ben Jehuda, R.; Shemer, Y.; Binah, O. Genome Editing in Induced Pluripotent Stem Cells using CRISPR/Cas9. Stem Cell Rev. Rep. 2018, 14, 323–336. [Google Scholar] [CrossRef] [PubMed]
- Hockemeyer, D.; Jaenisch, R. Induced Pluripotent Stem Cells Meet Genome Editing. Cell Stem Cell 2016, 18, 573–586. [Google Scholar] [CrossRef]
- Xu, H.; Wang, B.; Ono, M.; Kagita, A.; Fujii, K.; Sasakawa, N.; Ueda, T.; Gee, P.; Nishikawa, M.; Nomura, M.; et al. Targeted Disruption of HLA Genes via CRISPR-Cas9 Generates iPSCs with Enhanced Immune Compatibility. Cell Stem Cell 2019, 24, 566–578.e567. [Google Scholar] [CrossRef]
- Saeedi, S.; Israel, S.; Nagy, C.; Turecki, G. The emerging role of exosomes in mental disorders. Transl. Psychiatry 2019, 9, 122. [Google Scholar] [CrossRef]
- Yang, H.; Feng, R.; Fu, Q.; Xu, S.; Hao, X.; Qiu, Y.; Feng, T.; Zeng, Z.; Chen, M.; Zhang, S. Human induced pluripotent stem cell-derived mesenchymal stem cells promote healing via TNF-α-stimulated gene-6 in inflammatory bowel disease models. Cell Death Dis. 2019, 10, 718. [Google Scholar] [CrossRef]
- Workman, M.J.; Troisi, E.; Targan, S.R.; Svendsen, C.N.; Barrett, R.J. Modeling Intestinal Epithelial Response to Interferon-γ in Induced Pluripotent Stem Cell-Derived Human Intestinal Organoids. Int. J. Mol. Sci. 2020, 22, 288. [Google Scholar] [CrossRef]
- Dai, S.; Mo, Y.; Wang, Y.; Xiang, B.; Liao, Q.; Zhou, M.; Li, X.; Li, Y.; Xiong, W.; Li, G.; et al. Chronic Stress Promotes Cancer Development. Front. Oncol. 2020, 10, 1492. [Google Scholar] [CrossRef]
- Eckerling, A.; Ricon-Becker, I.; Sorski, L.; Sandbank, E.; Ben-Eliyahu, S. Stress and cancer: Mechanisms, significance and future directions. Nat. Rev. Cancer 2021, 21, 767–785. [Google Scholar] [CrossRef] [PubMed]
- Katoh, M. Therapeutics targeting angiogenesis: Genetics and epigenetics, extracellular miRNAs and signaling networks (Review). Int. J. Mol. Med. 2013, 32, 763–767. [Google Scholar] [CrossRef] [PubMed]
- Cui, X.; Fu, Q.; Wang, X.; Xia, P.; Cui, X.; Bai, X.; Lu, Z. Molecular mechanisms and clinical applications of exosomes in prostate cancer. Biomark. Res. 2022, 10, 56. [Google Scholar] [CrossRef]
- Polanco, J.C.; Hand, G.R.; Briner, A.; Li, C.; Götz, J. Exosomes induce endolysosomal permeabilization as a gateway by which exosomal tau seeds escape into the cytosol. Acta Neuropathol. 2021, 141, 235–256. [Google Scholar] [CrossRef]
- Sridharan, B.; Lim, H.G. Exosomes and ultrasound: The future of theranostic applications. Mater. Today. Bio 2023, 19, 100556. [Google Scholar] [CrossRef] [PubMed]
- Fan, J.; Lee, C.S.; Kim, S.; Chen, C.; Aghaloo, T.; Lee, M. Generation of Small RNA-Modulated Exosome Mimetics for Bone Regeneration. ACS Nano 2020, 14, 11973–11984. [Google Scholar] [CrossRef] [PubMed]
Cell Sources | Characterization | Models | Therapeutic Effects | Cargos | Reference |
---|---|---|---|---|---|
miPSCs | TEM | MI | Mitigate cardiac remodeling and improve cardiac functions post myocardial infarction | [81] | |
miPSCs | WB (CD63, Tsg101) | MIRI | Prevent cardiomyocyte apoptosis in ischemic myocardium | miR21, HIF-1α-regulated miR210 | [82] |
miPSCs | EM, FCM, RT-PCR | MIRI | Improve LV function and enhance angiogenesis | global miRNA and proteomic profiling performed | [21] |
hiPSCs | FCM, BCA | MI | Reduce fibrosis in infarcted mice hearts | CD82 | [83] |
hiPSCs | TEM, NTA | MI | Facilitate cardiac repair through circulating miRNAs | circulating miRNAs | [84] |
hiPSCs | NTA, WB (CD63) | HF | Involved in the remodeling process and observed in primary cardiomyocytes | miRNA mRNA | [85] |
hiPSCs | TEM, WB (CD63, CD9) | Endothelial cell in vitro | Improve cardiac function and repair | miRNA | [86] |
hiPSCs | TEM, NTA | H9c2 in vitro | Protect against oxidative-stress-induced apoptosis | miRNA | [87] |
hiCMs | WB (CD63, CD81) | Dys-iCMs In vitro | Decrease reactive oxygen species and delay mitochondrial permeability | [88] | |
hiCMs | TEM, WB (CD63, CD81) | MI | Facilitate cardiac repair and avoid immune rejection | miRNA, LncRNA | [89] |
hiCMs | WB (CD81, CD63, flotillin-1, TSTG101) | MI | Improve recovery from myocardial infarction in swine | [12] | |
hiMSCs | EM, NanoFCM | MI | Promote cell viability through activating the Akt/Nrf2/HO-1 axis and improve cardiac function | [90] | |
hiMSCs | TEM, immunoblot | Rat skin wound model | Promote collagen synthesis and angiogenesis | [91] | |
hiMSCs | EM, NTA | HF | Improve cardiac function and increased EF relative to baseline values | miRNA | [92] |
hiMSCs | TEM, NTA, RT-PCR | Ischemic Adult Human Cardiomyocytes | Alter cardiac tissue-level remodeling | miR21-5p | [93] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Meng, W.-T.; Guo, H.-D. Small Extracellular Vesicles Derived from Induced Pluripotent Stem Cells in the Treatment of Myocardial Injury. Int. J. Mol. Sci. 2023, 24, 4577. https://doi.org/10.3390/ijms24054577
Meng W-T, Guo H-D. Small Extracellular Vesicles Derived from Induced Pluripotent Stem Cells in the Treatment of Myocardial Injury. International Journal of Molecular Sciences. 2023; 24(5):4577. https://doi.org/10.3390/ijms24054577
Chicago/Turabian StyleMeng, Wan-Ting, and Hai-Dong Guo. 2023. "Small Extracellular Vesicles Derived from Induced Pluripotent Stem Cells in the Treatment of Myocardial Injury" International Journal of Molecular Sciences 24, no. 5: 4577. https://doi.org/10.3390/ijms24054577
APA StyleMeng, W.-T., & Guo, H.-D. (2023). Small Extracellular Vesicles Derived from Induced Pluripotent Stem Cells in the Treatment of Myocardial Injury. International Journal of Molecular Sciences, 24(5), 4577. https://doi.org/10.3390/ijms24054577