Stem Cell-Derived Exosomes and Nanovesicles: Promotion of Cell Proliferation, Migration, and Anti-Senescence for Treatment of Wound Damage and Skin Ageing
Abstract
:1. Introduction
2. Somatic and Cancer Cell-Derived Exosomes
3. Cancer Stem Cell-Derived Exosomes
4. Multipotent Stem Cell-Derived Exosomes
5. Pluripotent Stem Cell (PSC) Derived Exosomes and CNVs
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Camussi, G.; Deregibus, M.C.; Bruno, S.; Cantaluppi, V.; Biancone, L. Exosomes/microvesicles as a mechanism of cell-to-cell communication. Kidney Int. 2010, 78, 838–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mateescu, B.; Kowal, E.J.; van Balkom, B.W.; Bartel, S.; Bhattacharyya, S.N.; Buzás, E.I.; Buck, A.H.; de Candia, P.; Chow, F.W.; Das, S. Obstacles and opportunities in the functional analysis of extracellular vesicle RNA–an ISEV position paper. J. Extracell. Vesicles 2017, 6, 1286095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Harding, C.; Heuser, J.; Stahl, P. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J. Cell Biol. 1983, 97, 329–339. [Google Scholar] [CrossRef]
- Ramirez, M.I.; Amorim, M.G.; Gadelha, C.; Milic, I.; Welsh, J.A.; Freitas, V.M.; Nawaz, M.; Akbar, N.; Couch, Y.; Makin, L. Technical challenges of working with extracellular vesicles. Nanoscale 2018, 10, 881–906. [Google Scholar] [CrossRef] [Green Version]
- Vlassov, A.V.; Magdaleno, S.; Setterquist, R.; Conrad, R. Exosomes: Current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim. Biophys. Acta Gen. Subj. 2012, 1820, 940–948. [Google Scholar] [CrossRef]
- Rani, S.; Ritter, T. The Exosome—A Naturally Secreted Nanoparticle and its Application to Wound Healing. Adv. Mater. 2016, 28, 5542–5552. [Google Scholar] [CrossRef]
- Pathan, M.; Fonseka, P.; Chitti, S.V.; Kang, T.; Sanwlani, R.; Van Deun, J.; Hendrix, A.; Mathivanan, S. Vesiclepedia 2019: A compendium of RNA, proteins, lipids and metabolites in extracellular vesicles. Nucleic Acids Res. 2018, 47, D516–D519. [Google Scholar] [CrossRef] [Green Version]
- Jang, S.C.; Kim, O.Y.; Yoon, C.M.; Choi, D.-S.; Roh, T.-Y.; Park, J.; Nilsson, J.; Lotvall, J.; Kim, Y.-K.; Gho, Y.S. Bioinspired exosome-mimetic nanovesicles for targeted delivery of chemotherapeutics to malignant tumors. ACS Nano 2013, 7, 7698–7710. [Google Scholar] [CrossRef]
- Howitt, J.; Hill, A.F. Exosomes in the pathology of neurodegenerative diseases. J. Biol. Chem. 2016, 291, 26589–26597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liang, X.; Ding, Y.; Zhang, Y.; Tse, H.-F.; Lian, Q. Paracrine mechanisms of mesenchymal stem cell-based therapy: Current status and perspectives. Cell Transplant. 2014, 23, 1045–1059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kraitchman, D.L.; Tatsumi, M.; Gilson, W.D.; Ishimori, T.; Kedziorek, D.; Walczak, P.; Segars, W.P.; Chen, H.H.; Fritzges, D.; Izbudak, I. Dynamic imaging of allogeneic mesenchymal stem cells trafficking to myocardial infarction. Circulation 2005, 112, 1451. [Google Scholar] [CrossRef] [Green Version]
- Toma, C.; Wagner, W.R.; Bowry, S.; Schwartz, A.; Villanueva, F. Fate of culture-expanded mesenchymal stem cells in the microvasculature: In vivo observations of cell kinetics. Circ. Res. 2009, 104, 398–402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Phinney, D.G.; Pittenger, M.F. Concise review: MSC-derived exosomes for cell-free therapy. Stem Cells 2017, 35, 851–858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kourembanas, S. Exosomes: Vehicles of intercellular signaling, biomarkers, and vectors of cell therapy. Annu. Rev. Physiol. 2015, 77, 13–27. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Z.G.; Buller, B.; Chopp, M. Exosomes—beyond stem cells for restorative therapy in stroke and neurological injury. Nat. Rev. Neurol. 2019, 15, 193–203. [Google Scholar] [CrossRef]
- Chen, L.; Tredget, E.E.; Wu, P.Y.; Wu, Y. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS ONE 2008, 3, e1886. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.W.; Fang, X.; Krasnodembskaya, A.; Howard, J.P.; Matthay, M.A. Concise review: Mesenchymal stem cells for acute lung injury: Role of paracrine soluble factors. Stem Cells 2011, 29, 913–919. [Google Scholar] [CrossRef] [Green Version]
- Huh, M.-I.; Kim, M.-S.; Kim, H.-K.; Lim, J.O. Effect of conditioned media collected from human amniotic fluid-derived stem cells (hAFSCs) on skin regeneration and photo-aging. J. Tissue Eng. Regen. Med. 2014, 11, 171–177. [Google Scholar] [CrossRef]
- Li, Q.; Chen, Y.; Ma, K.; Zhao, A.; Zhang, C.; Fu, X. Regenerative and reparative effects of human chorion-derived stem cell conditioned medium on photo-aged epidermal cells. Cell Cycle 2016, 15, 1144–1155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shim, J.H.; Park, J.-Y.; Lee, M.-G.; Kang, H.H.; Lee, T.R.; Shin, D.W. Human dermal stem/progenitor cell-derived conditioned medium ameliorates ultraviolet a-induced damage of normal human dermal fibroblasts. PLoS ONE 2013, 8, e67604. [Google Scholar] [CrossRef] [PubMed]
- Jung, J.-Y.; Shim, J.H.; Choi, H.; Lee, T.R.; Shin, D.W. Human dermal stem/progenitor cell-derived conditioned medium improves senescent human dermal fibroblasts. Int. J. Mol. Sci. 2015, 16, 19027–19039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fatima, F.; Ekstrom, K.; Nazarenko, I.; Maugeri, M.; Valadi, H.; Hill, A.F.; Camussi, G.; Nawaz, M. Non-coding RNAs in mesenchymal stem cell-derived extracellular vesicles: Deciphering regulatory roles in stem cell potency, inflammatory resolve, and tissue regeneration. Front. Genet. 2017, 8, 161. [Google Scholar] [CrossRef] [Green Version]
- Nawaz, M.; Fatima, F.; Vallabhaneni, K.C.; Penfornis, P.; Valadi, H.; Ekström, K.; Kholia, S.; Whitt, J.D.; Fernandes, J.D.; Pochampally, R. Extracellular vesicles: Evolving factors in stem cell biology. Stem Cells Int. 2016, 2016, 1073140. [Google Scholar] [CrossRef] [Green Version]
- Falanga, A.; Marchetti, M.; Vignoli, A.; Balducci, D. Clotting mechanisms and cancer: Implications in thrombus formation and tumor progression. Clin. Adv. Hematol. Oncol. 2003, 1, 673–678. [Google Scholar]
- Janowska-Wieczorek, A.; Wysoczynski, M.; Kijowski, J.; Marquez-Curtis, L.; Machalinski, B.; Ratajczak, J.; Ratajczak, M.Z. Microvesicles derived from activated platelets induce metastasis and angiogenesis in lung cancer. Int. J. Cancer 2005, 113, 752–760. [Google Scholar] [CrossRef]
- Gutkin, A.; Uziel, O.; Beery, E.; Nordenberg, J.; Pinchasi, M.; Goldvaser, H.; Henick, S.; Goldberg, M.; Lahav, M. Tumor cells derived exosomes contain hTERT mRNA and transform nonmalignant fibroblasts into telomerase positive cells. Oncotarget 2016, 7, 59173. [Google Scholar] [CrossRef] [Green Version]
- Takasugi, M.; Okada, R.; Takahashi, A.; Virya Chen, D.; Watanabe, S.; Hara, E. Small extracellular vesicles secreted from senescent cells promote cancer cell proliferation through EphA2. Nat. Commun. 2017, 8, 15729. [Google Scholar] [CrossRef]
- Huang, A.; Dong, J.; Li, S.; Wang, C.; Ding, H.; Li, H.; Su, X.; Ge, X.; Sun, L.; Bai, C. Exosomal transfer of vasorin expressed in hepatocellular carcinoma cells promotes migration of human umbilical vein endothelial cells. Int. J. Biol. Sci. 2015, 11, 961. [Google Scholar] [CrossRef] [PubMed]
- Vrijsen, K.; Sluijter, J.; Schuchardt, M.; Van Balkom, B.; Noort, W.; Chamuleau, S.; Doevendans, P. Cardiomyocyte progenitor cell-derived exosomes stimulate migration of endothelial cells. J. Cell. Mol. Med. 2010, 14, 1064–1070. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Munagala, R.; Aqil, F.; Jeyabalan, J.; Gupta, R.C. Bovine milk-derived exosomes for drug delivery. Cancer Lett. 2016, 371, 48–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sidhu, S.S.; Mengistab, A.T.; Tauscher, A.N.; LaVail, J.; Basbaum, C. The microvesicle as a vehicle for EMMPRIN in tumor–stromal interactions. Oncogene 2004, 23, 956–963. [Google Scholar] [CrossRef] [Green Version]
- Atay, S.; Gercel-Taylor, C.; Suttles, J.; Mor, G.; Taylor, D.D. Trophoblast-derived exosomes mediate monocyte recruitment and differentiation. Am. J. Reprod. Immunol. 2011, 65, 65–77. [Google Scholar] [CrossRef] [PubMed]
- Ma, T.; Chen, Y.; Chen, Y.; Meng, Q.; Sun, J.; Shao, L.; Yu, Y.; Huang, H.; Hu, Y.; Yang, Z. MicroRNA-132, delivered by mesenchymal stem cell-derived exosomes, promote angiogenesis in myocardial infarction. Stem Cells Int. 2018, 2018, 3290372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, W.; Huang, L.; Li, Y.; Zhang, X.; Gu, J.; Yan, Y.; Xu, X.; Wang, M.; Qian, H.; Xu, W. Exosomes derived from human bone marrow mesenchymal stem cells promote tumor growth in vivo. Cancer Lett. 2012, 315, 28–37. [Google Scholar] [CrossRef]
- Cantaluppi, V.; Biancone, L.; Figliolini, F.; Beltramo, S.; Medica, D.; Deregibus, M.C.; Galimi, F.; Romagnoli, R.; Salizzoni, M.; Tetta, C. Microvesicles derived from endothelial progenitor cells enhance neoangiogenesis of human pancreatic islets. Cell Transplant. 2012, 21, 1305–1320. [Google Scholar] [CrossRef]
- Skog, J.; Würdinger, T.; Van Rijn, S.; Meijer, D.H.; Gainche, L.; Curry, W.T.; Carter, B.S.; Krichevsky, A.M.; Breakefield, X.O. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat. Cell Biol. 2008, 10, 1470–1476. [Google Scholar] [CrossRef]
- Yang, L.; Wu, X.-H.; Wang, D.; Luo, C.-L.; Chen, L.-X. Bladder cancer cell-derived exosomes inhibit tumor cell apoptosis and induce cell proliferation in vitro. Mol. Med. Rep. 2013, 8, 1272–1278. [Google Scholar] [CrossRef] [Green Version]
- Matsumoto, A.; Takahashi, Y.; Nishikawa, M.; Sano, K.; Morishita, M.; Charoenviriyakul, C.; Saji, H.; Takakura, Y. Accelerated growth of B16 BL 6 tumor in mice through efficient uptake of their own exosomes by B16 BL 6 cells. Cancer Sci. 2017, 108, 1803–1810. [Google Scholar] [CrossRef] [PubMed]
- Goulet, C.R.; Champagne, A.; Bernard, G.; Vandal, D.; Chabaud, S.; Pouliot, F.; Bolduc, S. Cancer-associated fibroblasts induce epithelial–mesenchymal transition of bladder cancer cells through paracrine IL-6 signalling. BMC Cancer 2019, 19, 137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, J.; Yu, J.; Zhang, H.; Wang, B.; Guo, H.; Bai, J.; Wang, J.; Dong, Y.; Zhao, Y.; Wang, Y. Exosomes-derived MiR-302b suppresses lung cancer cell proliferation and migration via TGFβRII inhibition. Cell. Physiol. Biochem. 2016, 38, 1715–1726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ramteke, A.; Ting, H.; Agarwal, C.; Mateen, S.; Somasagara, R.; Hussain, A.; Graner, M.; Frederick, B.; Agarwal, R.; Deep, G. Exosomes secreted under hypoxia enhance invasiveness and stemness of prostate cancer cells by targeting adherens junction molecules. Mol. Carcinog. 2015, 54, 554–565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Richards, K.E.; Zeleniak, A.E.; Fishel, M.L.; Wu, J.; Littlepage, L.E.; Hill, R. Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene 2017, 36, 1770–1778. [Google Scholar] [CrossRef] [Green Version]
- Huang, Z.; Feng, Y. Exosomes derived from hypoxic colorectal cancer cells promote angiogenesis through Wnt4-induced β-catenin signaling in endothelial cells. Oncol. Res. 2017, 25, 651–661. [Google Scholar] [CrossRef]
- Qu, J.-L.; Qu, X.-J.; Zhao, M.-F.; Teng, Y.-E.; Zhang, Y.; Hou, K.-Z.; Jiang, Y.-H.; Yang, X.-H.; Liu, Y.-P. Gastric cancer exosomes promote tumour cell proliferation through PI3K/Akt and MAPK/ERK activation. Dig. Liver Dis. 2009, 41, 875–880. [Google Scholar] [CrossRef]
- Abusamra, A.J.; Zhong, Z.; Zheng, X.; Li, M.; Ichim, T.E.; Chin, J.L.; Min, W.-P. Tumor exosomes expressing Fas ligand mediate CD8+ T-cell apoptosis. Blood Cells Mol. Dis. 2005, 35, 169–173. [Google Scholar] [CrossRef]
- Wu, L.; Zhang, X.; Zhang, B.; Shi, H.; Yuan, X.; Sun, Y.; Pan, Z.; Qian, H.; Xu, W. Exosomes derived from gastric cancer cells activate NF-κB pathway in macrophages to promote cancer progression. Tumor Biol. 2016, 37, 12169–12180. [Google Scholar] [CrossRef]
- Gutzeit, C.; Nagy, N.; Gentile, M.; Lyberg, K.; Gumz, J.; Vallhov, H.; Puga, I.; Klein, E.; Gabrielsson, S.; Cerutti, A.; et al. Exosomes Derived from Burkitt’s Lymphoma Cell Lines Induce Proliferation, Differentiation, and Class-Switch Recombination in B Cells. J. Immunol. 2014, 192, 5852–5862. [Google Scholar] [CrossRef] [Green Version]
- Jahanban-Esfahlan, A.; Dastmalchi, S.; Davaran, S. A simple improved desolvation method for the rapid preparation of albumin nanoparticles. Int. J. Biol. Macromol. 2016, 91, 703–709. [Google Scholar] [CrossRef] [PubMed]
- Shimbo, K.; Miyaki, S.; Ishitobi, H.; Kato, Y.; Kubo, T.; Shimose, S.; Ochi, M. Exosome-formed synthetic microRNA-143 is transferred to osteosarcoma cells and inhibits their migration. Biochem. Biophys. Res. Commun. 2014, 445, 381–387. [Google Scholar] [CrossRef] [PubMed]
- Adams, R.H.; Alitalo, K. Molecular regulation of angiogenesis and lymphangiogenesis. Nat. Rev. Mol. 2007, 8, 464–478. [Google Scholar] [CrossRef]
- Bazigou, E.; Makinen, T. Flow control in our vessels: Vascular valves make sure there is no way back. Cell. Mol. Life Sci. 2013, 70, 1055–1066. [Google Scholar] [CrossRef] [Green Version]
- Hood, J.L.; Pan, H.; Lanza, G.M.; Wickline, S.A. Paracrine induction of endothelium by tumor exosomes. Lab. Investig. 2009, 89, 1317–1328. [Google Scholar] [CrossRef] [Green Version]
- Hood, J.L.; San, R.S.; Wickline, S.A. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 2011, 71, 3792–3801. [Google Scholar] [CrossRef] [Green Version]
- Kucharzewska, P.; Christianson, H.C.; Welch, J.E.; Svensson, K.J.; Fredlund, E.; Ringnér, M.; Mörgelin, M.; Bourseau-Guilmain, E.; Bengzon, J.; Belting, M. Exosomes reflect the hypoxic status of glioma cells and mediate hypoxia-dependent activation of vascular cells during tumor development. Proc. Natl. Acad. Sci. USA 2013, 110, 7312–7317. [Google Scholar] [CrossRef] [Green Version]
- Zhou, W.; Fong, M.Y.; Min, Y.; Somlo, G.; Liu, L.; Palomares, M.R.; Yu, Y.; Chow, A.; O’Connor, S.T.F.; Chin, A.R. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell 2014, 25, 501–515. [Google Scholar] [CrossRef] [Green Version]
- Ngalame, N.N.; Luz, A.L.; Makia, N.; Tokar, E.J. Arsenic alters exosome quantity and cargo to mediate stem cell recruitment into a cancer stem cell-like phenotype. Toxicol. Sci. 2018, 165, 40–49. [Google Scholar] [CrossRef] [Green Version]
- Ma, Z.; Cui, X.; Lu, L.; Chen, G.; Yang, Y.; Hu, Y.; Lu, Y.; Cao, Z.; Wang, Y.; Wang, X. Exosomes from glioma cells induce a tumor-like phenotype in mesenchymal stem cells by activating glycolysis. Stem Cell. Res. Ther. 2019, 10, 1–18. [Google Scholar] [CrossRef] [Green Version]
- Ayob, A.Z.; Ramasamy, T.S. Cancer stem cells as key drivers of tumour progression. J. Biomed. Sci. 2018, 25, 1–18. [Google Scholar] [CrossRef] [PubMed]
- Ribatti, D. Cancer stem cells and tumor angiogenesis. Cancer Lett. 2012, 321, 13–17. [Google Scholar] [CrossRef] [PubMed]
- Sugimori, M.; Hayakawa, Y.; Boman, B.M.; Fields, J.Z.; Awaji, M.; Kozano, H.; Tamura, R.; Yamamoto, S.; Ogata, T.; Yamada, M. Discovery of power-law growth in the self-renewal of heterogeneous glioma stem cell populations. PLoS ONE 2015, 10, e0135760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, X.; Ma, X.; Wang, J.; Zhao, Y.; Wang, Y.; Bihl, J.C.; Chen, Y.; Jiang, C. Glioma stem cells-derived exosomes promote the angiogenic ability of endothelial cells through miR-21/VEGF signal. Oncotarget 2017, 8, 36137. [Google Scholar] [CrossRef] [Green Version]
- Grange, C.; Tapparo, M.; Collino, F.; Vitillo, L.; Damasco, C.; Deregibus, M.C.; Tetta, C.; Bussolati, B.; Camussi, G. Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Cancer Res. 2011, 71, 5346–5356. [Google Scholar] [CrossRef] [Green Version]
- Conigliaro, A.; Costa, V.; Dico, A.L.; Saieva, L.; Buccheri, S.; Dieli, F.; Manno, M.; Raccosta, S.; Mancone, C.; Tripodi, M. CD90+ liver cancer cells modulate endothelial cell phenotype through the release of exosomes containing H19 lncRNA. Mol. Cancer 2015, 14, 155. [Google Scholar] [CrossRef]
- Al-Sowayan, B.S.; Al-Shareeda, A.T.; Alrfaei, B.M. Cancer Stem Cell-Exosomes, Unexposed Player in Tumorigenicity. Front. Pharmacol. 2020, 11, 384. [Google Scholar] [CrossRef] [Green Version]
- Sánchez, C.A.; Andahur, E.I.; Valenzuela, R.; Castellón, E.A.; Fullá, J.A.; Ramos, C.G.; Triviño, J.C. Exosomes from bulk and stem cells from human prostate cancer have a differential microRNA content that contributes cooperatively over local and pre-metastatic niche. Oncotarget 2016, 7, 3993. [Google Scholar] [CrossRef] [Green Version]
- Gabrusiewicz, K.; Li, X.; Wei, J.; Hashimoto, Y.; Marisetty, A.L.; Ott, M.; Wang, F.; Hawke, D.; Yu, J.; Healy, L.M. Glioblastoma stem cell-derived exosomes induce M2 macrophages and PD-L1 expression on human monocytes. Oncoimmunology 2018, 7, e1412909. [Google Scholar] [CrossRef]
- Sun, Z.-P.; Li, A.-Q.; Jia, W.-H.; Ye, S.; Van Eps, G.; Yu, J.-M.; Yang, W.-J. MicroRNA expression profiling in exosomes derived from gastric cancer stem-like cells. Oncotarget 2017, 8, 93839. [Google Scholar] [CrossRef] [Green Version]
- Hardin, H.; Helein, H.; Meyer, K.; Robertson, S.; Zhang, R.; Zhong, W.; Lloyd, R.V. Thyroid cancer stem-like cell exosomes: Regulation of EMT via transfer of lncRNAs. Lab. Investig. 2018, 98, 1133–1142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, L.; Yang, G.; Zhao, D.; Wang, J.; Bai, Y.; Peng, Q.; Wang, H.; Fang, R.; Chen, G.; Wang, Z. CD103-positive CSC exosome promotes EMT of clear cell renal cell carcinoma: Role of remote MiR-19b-3p. Mol. Cancer 2019, 18, 86. [Google Scholar] [CrossRef]
- Hwang, W.-L.; Lan, H.-Y.; Cheng, W.-C.; Huang, S.-C.; Yang, M.-H. Tumor stem-like cell-derived exosomal RNAs prime neutrophils for facilitating tumorigenesis of colon cancer. J. Hematol. Oncol. 2019, 12, 1–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ciardiello, C.; Leone, A.; Budillon, A. The crosstalk between cancer stem cells and microenvironment is critical for solid tumor progression: The significant contribution of extracellular vesicles. Stem Cells Int. 2018, 2018, 6392198. [Google Scholar] [CrossRef] [PubMed]
- Rashed, M.H.; Bayraktar, E.; Helal, G.K.; Abd-Ellah, M.F.; Amero, P.; Chavez-Reyes, A.; Rodriguez-Aguayo, C. Exosomes: From garbage bins to promising therapeutic targets. Int. J. Mol. Sci. 2017, 18, 538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barry, F.P.; Murphy, J.M. Mesenchymal stem cells: Clinical applications and biological characterization. Int. J. Biochem. Cell Biol. 2004, 36, 568–584. [Google Scholar] [CrossRef]
- Krampera, M.; Pizzolo, G.; Aprili, G.; Franchini, M. Mesenchymal stem cells for bone, cartilage, tendon and skeletal muscle repair. Bone 2006, 39, 678–683. [Google Scholar] [CrossRef] [PubMed]
- Choi, D.S.; Park, J.O.; Jang, S.C.; Yoon, Y.J.; Jung, J.W.; Choi, D.Y.; Kim, J.W.; Kang, J.S.; Park, J.; Hwang, D. Proteomic analysis of microvesicles derived from human colorectal cancer ascites. Proteomics 2011, 11, 2745–2751. [Google Scholar] [CrossRef]
- Anderson, J.D.; Johansson, H.J.; Graham, C.S.; Vesterlund, M.; Pham, M.T.; Bramlett, C.S.; Montgomery, E.N.; Mellema, M.S.; Bardini, R.L.; Contreras, Z. Comprehensive proteomic analysis of mesenchymal stem cell exosomes reveals modulation of angiogenesis via nuclear factor-kappaB signaling. Stem Cells 2016, 34, 601–613. [Google Scholar] [CrossRef] [Green Version]
- Vallabhaneni, K.C.; Hassler, M.-Y.; Abraham, A.; Whitt, J.; Mo, Y.-Y.; Atfi, A.; Pochampally, R. Mesenchymal stem/stromal cells under stress increase osteosarcoma migration and apoptosis resistance via extracellular vesicle mediated communication. PLoS ONE 2016, 11, e0166027. [Google Scholar] [CrossRef]
- Shabbir, A.; Cox, A.; Rodriguez-Menocal, L.; Salgado, M.; Badiavas, E.V. Mesenchymal stem cell exosomes induce proliferation and migration of normal and chronic wound fibroblasts, and enhance angiogenesis in vitro. Stem Cells Dev. 2015, 24, 1635–1647. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Yoo, J.K.; Kim, J.; Choi, S.J.; Noh, H.M.; Kwon, Y.D.; Yoo, H.; Yi, H.s.; Chung, H.M.; Kim, J.K. Discovery and characterization of novel microRNAs during endothelial differentiation of human embryonic stem cells. Stem Cells Dev. 2012, 21, 2049–2057. [Google Scholar] [CrossRef] [Green Version]
- Zhang, B.; Wang, M.; Gong, A.; Zhang, X.; Wu, X.; Zhu, Y.; Shi, H.; Wu, L.; Zhu, W.; Qian, H. HucMSC-exosome mediated-Wnt4 signaling is required for cutaneous wound healing. Stem Cells 2015, 33, 2158–2168. [Google Scholar] [CrossRef] [PubMed]
- Nong, K.; Wang, W.; Niu, X.; Hu, B.; Ma, C.; Bai, Y.; Wu, B.; Wang, Y.; Ai, K. Hepatoprotective effect of exosomes from human-induced pluripotent stem cell–derived mesenchymal stromal cells against hepatic ischemia-reperfusion injury in rats. Cytotherapy 2016, 18, 1548–1559. [Google Scholar] [CrossRef] [PubMed]
- Du, Y.; Li, D.; Han, C.; Wu, H.; Xu, L.; Zhang, M.; Zhang, J.; Chen, X. Exosomes from human-induced pluripotent stem cell–derived mesenchymal stromal cells (hiPSC-MSCs) protect liver against hepatic ischemia/reperfusion injury via activating sphingosine kinase and sphingosine-1-phosphate signaling pathway. Cell. Physiol. Biochem. 2017, 43, 611–625. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Nakamura, Y.; Miyaki, S.; Ishitobi, H.; Matsuyama, S.; Nakasa, T.; Kamei, N.; Akimoto, T.; Higashi, Y.; Ochi, M. Mesenchymal-stem-cell-derived exosomes accelerate skeletal muscle regeneration. FEBS Lett. 2015, 589, 1257–1265. [Google Scholar] [CrossRef] [Green Version]
- Chen, B.; Li, Q.; Zhao, B.; Wang, Y. Stem cell-derived extracellular vesicles as a novel potential therapeutic tool for tissue repair. Stem Cells Transl. Med. 2017, 6, 1753–1758. [Google Scholar] [CrossRef]
- Goodarzi, P.; Larijani, B.; Alavi-Moghadam, S.; Tayanloo-Beik, A.; Mohamadi-Jahani, F.; Ranjbaran, N.; Payab, M.; Falahzadeh, K.; Mousavi, M.; Arjmand, B. Mesenchymal stem cells-derived exosomes for wound regeneration. In Cell Biology and Translational Medicine; Springer: Berlin/Heidelberg, Germany, 2018; Volume 4, pp. 119–131. [Google Scholar]
- Maxson, S.; Lopez, E.A.; Yoo, D.; Danilkovitch-Miagkova, A.; LeRoux, M.A. Concise review: Role of mesenchymal stem cells in wound repair. Stem Cells Transl. Med. 2012, 1, 142–149. [Google Scholar] [CrossRef]
- Phinney, D.G.; Prockop, D.J. Concise review: Mesenchymal stem/multipotent stromal cells: The state of transdifferentiation and modes of tissue repair—Current views. Stem Cells 2007, 25, 2896–2902. [Google Scholar] [CrossRef] [PubMed]
- Wu, Y.; Wang, J.; Scott, P.G.; Tredget, E.E. Bone marrow-derived stem cells in wound healing: A review. Wound Repair Regen. 2007, 15, S18–S26. [Google Scholar] [CrossRef] [PubMed]
- Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [Green Version]
- Hu, L.; Wang, J.; Zhou, X.; Xiong, Z.; Zhao, J.; Yu, R.; Huang, F.; Zhang, H.; Chen, L. Exosomes derived from human adipose mensenchymal stem cells accelerates cutaneous wound healing via optimizing the characteristics of fibroblasts. Sci. Rep. 2016, 6, 32993. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.; Lee, S.K.; Kim, H.; Kim, T.M. Exosomes secreted from induced pluripotent stem cell-derived mesenchymal stem cells accelerate skin cell proliferation. Int. J. Mol. Sci. 2018, 19, 3119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Han, C.; Jeong, D.; Kim, B.; Jo, W.; Kang, H.; Cho, S.; Kim, K.H.; Park, J. Mesenchymal Stem Cell Engineered Nanovesicles for Accelerated Skin Wound Closure. ACS Biomater. Sci. Eng. 2019, 5, 1534–1543. [Google Scholar] [CrossRef]
- Ma, Y.; Ge, S.; Zhang, J.; Zhou, D.; Li, L.; Wang, X.; Su, J. Mesenchymal stem cell-derived extracellular vesicles promote nerve regeneration after sciatic nerve crush injury in rats. Int. J. Clin. Exp. Pathol. 2017, 10, 10032. [Google Scholar]
- Ma, Y.; Dong, L.; Zhou, D.; Li, L.; Zhang, W.; Zhen, Y.; Wang, T.; Su, J.; Chen, D.; Mao, C. Extracellular vesicles from human umbilical cord mesenchymal stem cells improve nerve regeneration after sciatic nerve transection in rats. J. Cell. Mol. Med. 2019, 23, 2822–2835. [Google Scholar] [CrossRef] [Green Version]
- Figley, S.A.; Khosravi, R.; Legasto, J.M.; Tseng, Y.-F.; Fehlings, M.G. Characterization of vascular disruption and blood–spinal cord barrier permeability following traumatic spinal cord injury. J. Neurotrauma 2014, 31, 541–552. [Google Scholar] [CrossRef] [Green Version]
- Lu, Y.; Zhou, Y.; Zhang, R.; Wen, L.; Wu, K.; Li, Y.; Yao, Y.; Duan, R.; Jia, Y. Bone mesenchymal stem cell-derived extracellular vesicles promote recovery following spinal cord injury via improvement of the integrity of the blood-spinal cord barrier. Front. Neurosci. 2019, 13, e0166027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, C.; Jiao, G.; Wu, W.; Wang, H.; Ren, S.; Zhang, L.; Zhou, H.; Liu, H.; Chen, Y. Exosomes from Bone Marrow Mesenchymal Stem Cells Inhibit Neuronal Apoptosis and Promote Motor Function Recovery via the Wnt/β-catenin Signaling Pathway. Cell Transplant. 2019, 28, 1373–1383. [Google Scholar] [CrossRef] [PubMed]
- Rong, Y.; Liu, W.; Wang, J.; Fan, J.; Luo, Y.; Li, L.; Kong, F.; Chen, J.; Tang, P.; Cai, W. Neural stem cell-derived small extracellular vesicles attenuate apoptosis and neuroinflammation after traumatic spinal cord injury by activating autophagy. Cell Death Dis. 2019, 10, 1–18. [Google Scholar] [CrossRef] [PubMed]
- Lelek, J.; Zuba-Surma, E.K. Perspectives for Future Use of Extracellular Vesicles from Umbilical Cord-and Adipose Tissue-Derived Mesenchymal Stem/Stromal Cells in Regenerative Therapies—Synthetic Review. Int. J. Mol. Sci. 2020, 21, 799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khan, M.; Nickoloff, E.; Abramova, T.; Johnson, J.; Verma, S.K.; Krishnamurthy, P.; Mackie, A.R.; Vaughan, E.; Garikipati, V.N.S.; Benedict, C. Embryonic stem cell–derived exosomes promote endogenous repair mechanisms and enhance cardiac function following myocardial infarction. Circ. Res. 2015, 117, 52–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Povero, D.; Pinatel, E.M.; Leszczynska, A.; Goyal, N.P.; Nishio, T.; Kim, J.; Kneiber, D.; de Araujo Horcel, L.; Eguchi, A.; Ordonez, P.M. Human induced pluripotent stem cell–derived extracellular vesicles reduce hepatic stellate cell activation and liver fibrosis. JCI Insight 2019, 4, e125652. [Google Scholar] [CrossRef]
- Oh, M.; Kim, Y.J.; Son, Y.J.; Yoo, H.S.; Park, J.H. Promotive effects of human induced pluripotent stem cell-conditioned medium on the proliferation and migration of dermal fibroblasts. Biotechnol. Bioprocess Eng. 2017, 22, 561–568. [Google Scholar] [CrossRef]
- Oh, M.; Lee, J.; Kim, Y.J.; Rhee, W.J.; Park, J.H. Exosomes derived from human induced pluripotent stem cells ameliorate the aging of skin fibroblasts. Int. J. Mol. Sci. 2018, 19, 1715. [Google Scholar] [CrossRef] [Green Version]
- Branscome, H.; Paul, S.; Khatkar, P.; Kim, Y.; Barclay, R.A.; Pinto, D.O.; Yin, D.; Zhou, W.; Liotta, L.A.; El-Hage, N. Stem cell extracellular vesicles and their potential to contribute to the repair of damaged CNS cells. J. Neuroimmune Pharmacol. 2020, 15, 520–537. [Google Scholar] [CrossRef]
- Liu, S.; Mahairaki, V.; Bai, H.; Ding, Z.; Li, J.; Witwer, K.W.; Cheng, L. Highly purified human extracellular vesicles produced by stem cells alleviate aging cellular phenotypes of senescent human cells. Stem Cells 2019, 37, 779–790. [Google Scholar] [CrossRef] [Green Version]
- Kim, J.; Han, C.; Jo, W.; Kang, S.; Cho, S.; Jeong, D.; Gho, Y.S.; Park, J. Cell-Engineered Nanovesicle as a Surrogate Inducer of Contact-Dependent Stimuli. Adv. Healthc. Mater. 2017, 6, 1700381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, H.; Cha, H.; Park, J.H. Derivation of Cell-Engineered Nanovesicles from Human Induced Pluripotent Stem Cells and Their Protective Effect on the Senescence of Dermal Fibroblasts. Int. J. Mol. Sci. 2020, 21, 343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van Deun, J.; Mestdagh, P.; Sormunen, R.; Cocquyt, V.; Vermaelen, K.; Vandesompele, J.; Bracke, M.; De Wever, O.; Hendrix, A. The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling. J. Extracell. Vesicles 2014, 3, 24858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jo, W.; Jeong, D.; Kim, J.; Park, J. Self-Renewal of Bone Marrow Stem Cells by Nanovesicles Engineered from Embryonic Stem Cells. Adv. Healthc. Mater. 2016, 5, 3148–3156. [Google Scholar] [CrossRef] [PubMed]
- Jeong, D.; Jo, W.; Yoon, J.; Kim, J.; Gianchandani, S.; Gho, Y.S.; Park, J. Nanovesicles engineered from ES cells for enhanced cell proliferation. Biomaterials 2014, 35, 9302–9310. [Google Scholar] [CrossRef] [PubMed]
EV Donor Stem Cells | EVs | EV Recipient Cells/In Vivo Model | Effect | Ref |
---|---|---|---|---|
Human glioma stem cells (U-251 cell line) | Exosomes | Endothelial cells (Human brain ECs) | Angiogenesis | [64] |
Human liver cancer stem cells (Huh-7 human hepatocellular carcinoma cell line) | Exosomes | Endothelial cells (HUVEC) | Angiogenesis | [66] |
Human renal cancer stem cells (5 Patients) | MVs | Endothelial cells (HUVEC)/lung premetastic niche in SCID mice In vivo | Angiogenesis | [65] |
Human glioblastoma stem cell | Exosomes | Human monocytes | Induce M2 macrophages | [69] |
Human clear cell renal cell carcinoma stem cell | Exosomes | Clear cell renal cell carcinoma in BALB/c nude mice In vivo | Promote epithelial-mesenchymal transition | [71] |
Human thyroid cancer stem cell | Exosomes | Thyroid papillary carcinoma cell line (TPC-1)/Normal thyroid cell line (NTHY-ori-3) | Induce epithelial-mesenchymal transition | [72] |
EV Donor Stem Cells | EVs | EV Recipient Cells/In Vivo Model | Effect | Ref |
---|---|---|---|---|
Human BM-MSCs | Exosomes | HUVEC, mouse C2C12 myoblast cell line/cardiotoxin muscle injury model in mice | Promote myogenesis and angiogenesis in vitro/Promote muscle regeneration in a muscle injury model | [88] |
Human iPSC-MSCs | Exosomes | Injected systemically via the inferior vena cava in a rat model | Alleviate hepatic ischemia-reperfusion injury in rats (lowered Hepatocyte injury markers (AST and ALT) and inflammatory markers (TNF-α, IL-6, HMGB1)) | [85] |
Human iPSC-MSCs | Exosomes | Injected systemically into a murine ischemia/reperfusion injury model via the inferior vena cava | Induce primary hepatocytes and HL-7702 cells proliferation in vitro/Induce expression levels of proliferation markers (PCNA and PHH3) in vivo | [86] |
Human iPSC-MSCs | Exosomes | Bone marrow MSCs derived from female ovariectomized rats (rBMSCs-OVX) in vitro/implanted into bone defects in ovariectomized rats | Repair bone defects via increased angiogenesis and osteogenesis in osteoporotic rats | [87] |
Human iPSC-MSCs | Exosomes | HDFs and HUVECs/injected subcutaneously around wound sites in a rat model | Stimulate proliferation and migration/Accelerate re-epithelialization, facilitate angiogenesis and cutaneous wound healing in vivo | [95] |
Human adiopose-MSCs | Exosomes | Primary HDFs/injected intravenously | Promote migration, proliferation, collagen synthesis/Accelerate cutaneous wound healing in vivo | [96] |
Mouse BM-MSCs | EVs | Sciatic nerve crushed rat model | Promote peripheral nerve regeneration | [99] |
Mouse UC-MSCs | EVs | Left sciatic nerves removed rat model | Promote peripheral nerve regeneration | [100] |
Mouse BM-MSCs | EVs | SCI rat model | Improving the structural integrity of BSCB | [102] |
Mouse BM-MSCs | EVs | Neuronal cells/SCI rat model | Anti-apoptosis of neuron cells | [103] |
Mouse neural stem cell | EVs | Murine spinal neuron/rat model of SCI | Attenuate apoptosis and neuroinflammation | [104] |
Human iPSC-MSCs | Exosomes | Human HaCaT keratinocytes and HDFs | Accelerate skin cell proliferation | [97] |
Mouse MSCs | CNVs | Primary mouse skin fibroblasts/Injected IP into mouse skin wound model | Increase proliferation and migration in vitro/Accelerate healing in vivo | [98] |
EV Donor Stem Cells | EVs | EV Recipient Cells/In Vivo Model | Effect | Ref |
---|---|---|---|---|
Mouse ESCs | Exosomes | H9c2 myoblasts and HUVECs, cardiac progenitor cells (CPCs) | Enhance tube formation in vitro/Promote endogenous repair (augment neovascularization, myocyte proliferation, and survival) and enhance cardiac function in vivo | [106] |
Murine ESCs | CNVs | Primary murine skin fibroblasts | Increase proliferation rate and growth factor secretion | [116] |
Human iPSCs | Exosomes | HDFs | Stimulate proliferation, migration, and reduce photoaging, natural senescence markers (ameliorate skin ageing) | [109] |
Human iPSCs | EVs | Senescent human MSCs | Improve proliferation, reduce intracellular reactive oxygen species (ROS) level (alleviate aging) | [111] |
Mouse ESCs | CNVs | Murine MSCs | Enhance the proliferation rate | [115] |
Human iPSCs | CNVs | Senescent human HDFs | Increase proliferation, migration, and reduce activity of senescence-associated genes | [113] |
Human iPSCs | EVs | Hepatic stellate cell | Reduce hepatic stellate cell activation and liver fibrosis | [107] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Cha, H.; Hong, S.; Park, J.H.; Park, H.H. Stem Cell-Derived Exosomes and Nanovesicles: Promotion of Cell Proliferation, Migration, and Anti-Senescence for Treatment of Wound Damage and Skin Ageing. Pharmaceutics 2020, 12, 1135. https://doi.org/10.3390/pharmaceutics12121135
Cha H, Hong S, Park JH, Park HH. Stem Cell-Derived Exosomes and Nanovesicles: Promotion of Cell Proliferation, Migration, and Anti-Senescence for Treatment of Wound Damage and Skin Ageing. Pharmaceutics. 2020; 12(12):1135. https://doi.org/10.3390/pharmaceutics12121135
Chicago/Turabian StyleCha, Hyeonjin, Seyoung Hong, Ju Hyun Park, and Hee Ho Park. 2020. "Stem Cell-Derived Exosomes and Nanovesicles: Promotion of Cell Proliferation, Migration, and Anti-Senescence for Treatment of Wound Damage and Skin Ageing" Pharmaceutics 12, no. 12: 1135. https://doi.org/10.3390/pharmaceutics12121135
APA StyleCha, H., Hong, S., Park, J. H., & Park, H. H. (2020). Stem Cell-Derived Exosomes and Nanovesicles: Promotion of Cell Proliferation, Migration, and Anti-Senescence for Treatment of Wound Damage and Skin Ageing. Pharmaceutics, 12(12), 1135. https://doi.org/10.3390/pharmaceutics12121135