The Regenerative Role of the Fetal and Adult Stem Cell Secretome

Sveva Bollini, Chiara Gentili, Roberta Tasso, Ranieri Cancedda, Sveva Bollini, Chiara Gentili, Roberta Tasso, Ranieri Cancedda

Abstract

For a long time, the stem cell regenerative paradigm has been based on the assumption that progenitor cells play a critical role in tissue repair by means of their plasticity and differentiation potential. However, recent works suggest that the mechanism underlying the benefits of stem cell transplantation might relate to a paracrine modulatory effect rather than the replacement of affected cells at the site of injury. Therefore, mounting evidence that stem cells may act as a reservoir of trophic signals released to modulate the surrounding tissue has led to a paradigm shift in regenerative medicine. Attention has been shifted from analysis of the stem cell genome to understanding the stem cell "secretome", which is represented by the growth factors, cytokines and chemokines produced through paracrine secretion. Insights into paracrine-mediated repair support a new approach in regenerative medicine and the isolation and administration of specific stem cell-derived paracrine factors may represent an extremely promising strategy, introducing paracrine-based therapy as a novel and feasible clinical application. In this review, we will discuss the regenerative potential of fetal and adult stem cells, with particular attention to their secretome.

Keywords: microvesicles; paracrine effect; secretome; stem cells; tissue repair; trophic factors.

Figures

Figure 1
Figure 1
Schematic representation of paracrine therapy using adult or fetal mesenchymal stem cells. After isolation, stem cells are expanded in vitro and stimulated in order to release trophic factors into their conditioned medium via microvesicles/exosomes secretion. Factors directly released in the conditioned medium or contained in the microvesicles may be further isolated and use as therapeutic agent for tissue regeneration and repair.

References

    1. Caplan A.I., Dennis J.E. Mesenchymal stem cells as trophic mediators. J. Cell. Biochem. 2006;98:1076–1084. doi: 10.1002/jcb.20886.
    1. Kinnaird T., Stabile E., Burnett M.S., Lee C.W., Barr S., Fuchs S., Epstein S.E. Marrow-derived stromal cells express genes encoding a broad spectrum of arteriogenic cytokines and promote in vitro and in vivo arteriogenesis through paracrine mechanisms. Circ. Res. 2004;94:678–685.
    1. Mirotsou M., Jayawardena T.M., Schmeckpeper J., Gnecchi M., Dzau V.J. Paracrine mechanisms of stem cell reparative and regenerative actions in the heart. J. Mol. Cell. Cardiol. 2011;50:280–289. doi: 10.1016/j.yjmcc.2010.08.005.
    1. Cantaluppi V., Biancone L., Quercia A., Deregibus M.C., Segoloni G., Camussi G. Rationale of mesenchymal stem cell therapy in kidney injury. Am. J. Kidney Dis. 2013;61:300–309. doi: 10.1053/j.ajkd.2012.05.027.
    1. Kuo T.K., Hung S.P., Chuang C.H., Chen C.T., Shih Y.R., Fang S.C., Yang V.W., Lee O.K. Stem cell therapy for liver disease: Parameters governing the success of using bone marrow mesenchymal stem cells. Gastroenterology. 2008;134:2111–2121.
    1. 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. doi: 10.1002/stem.643.
    1. Uccelli A., Benvenuto F., Laroni A., Giunti D. Neuroprotective features of mesenchymal stem cells. Best Pract. Res. Clin. Haematol. 2011;24:59–64. doi: 10.1016/j.beha.2011.01.004.
    1. Mathivanan S., Fahner C.J., Reid G.E., Simpson R.J. ExoCarta 2012: Database of exosomal proteins, RNA and lipids. Nucleic Acids Res. 2012;40:1241–1244.
    1. Katsuda T., Kosaka N., Takeshita F., Ochiya T. The therapeutic potential of mesenchymal stem cell-derived extracellular vesicles. Proteomics. 2013;13:1637–1653. doi: 10.1002/pmic.201200373.
    1. Pan B.T., Johnstone R.M. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the receptor. Cell. 1983;33:967–978. doi: 10.1016/0092-8674(83)90040-5.
    1. Raposo G., Nijman H.W., Stoorvogel W., Liejendekker R., Harding C.V., Melief C.J., Geuze H.J. B lymphocytes secrete antigen-presenting vesicles. J. Exp. Med. 1996;183:1161–1172. doi: 10.1084/jem.183.3.1161.
    1. Lai R.C., Chen T.S., Lim S.K. Mesenchymal stem cell exosome: A novel stem cell-based therapy for cardiovascular disease. Regen. Med. 2011;6:481–492. doi: 10.2217/rme.11.35.
    1. Mathivanan S., Ji H., Simpson R.J. Exosomes: Extracellular organelles important in intercellular communication. J. Proteomics. 2010;73:1907–1920. doi: 10.1016/j.jprot.2010.06.006.
    1. Camussi G., Deregibus M.C., Cantaluppi V. Role of stem-cell-derived microvesicles in the paracrine action of stem cells. Biochem. Soc. Trans. 2013;41:283–287. doi: 10.1042/BST20120192.
    1. Verfaillie C.M. Adult stem cells: Assessing the case for pluripotency. Trends Cell Biol. 2002;12:502–508. doi: 10.1016/S0962-8924(02)02386-3.
    1. Thomson J.A., Itskovitz-Eldor J., Shapiro S.S., Waknitz M.A., Swiergiel J.J., Marshall V.S., Jones J.M. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282:1145–1147. doi: 10.1126/science.282.5391.1145.
    1. Pappa K.I., Anagnou N.P. Novel sources of fetal stem cells: Where do they fit on the developmental continuum? Regen. Med. 2009;4:423–433. doi: 10.2217/rme.09.12.
    1. Yen B.L., Huang H.I., Chien C.C., Jui H.Y., Ko B.S., Yao M., Shun C.T., Yen M.L., Lee M.C., Chen Y.C. Isolation of multipotent cells from human term placenta. Stem Cells. 2005;23:3–9. doi: 10.1634/stemcells.2004-0098.
    1. Miao Z., Jin J., Chen L., Zhu J., Huang W., Zhao J., Qian H., Zhang X. Isolation of mesenchymal stem cells from human placenta: Comparison with human bone marrow mesenchymal stem cells. Cell Biol. Int. 2006;30:681–687. doi: 10.1016/j.cellbi.2006.03.009.
    1. Ventura C., Cantoni S., Bianchi F., Lionetti V., Cavallini C., Scarlata I., Foroni L., Maioli M., Bonsi L., Alviano F., et al. Hyaluronan mixed esters of butyric and retinoic acid drive cardiac and endothelial fate in term placenta human mesenchymal stem cells and enhance cardiac repair in infarcted rat hearts. J. Biol. Chem. 2007;282:14243–14252. doi: 10.1074/jbc.M609350200.
    1. Chan J., Kennea N.L., Fisk N.M. Placental mesenchymal stem cells. Am. J. Obstet. Gynecol. 2007;196:e18. doi: 10.1016/j.ajog.2006.08.002.
    1. Wang M., Yang Y., Yang D., Luo F., Liang W., Guo S., Xu J. The immunomodulatory activity of human umbilical cord blood-derived mesenchymal stem cells in vitro. Immunology. 2009;126:220–232. doi: 10.1111/j.1365-2567.2008.02891.x.
    1. Liu S.H., Huang J.P., Lee R.K., Huang M.C., Wu Y.H., Chen C.Y., Chen C.P. Paracrine factors from human placental multipotent mesenchymal stromal cells protect endothelium from oxidative injury via STAT3 and manganese superoxide dismutase activation. Biol. Reprod. 2010;82:905–913. doi: 10.1095/biolreprod.109.081828.
    1. Kong P., Xie X., Li F., Liu Y., Lu Y. Placenta mesenchymal stem cell accelerates wound healing by enhancing angiogenesis in diabetic Goto-Kakizaki (GK) rats. Biochem. Biophys. Res. Commun. 2013;438:410–419. doi: 10.1016/j.bbrc.2013.07.088.
    1. Salomon C., Ryan J., Sobrevia L., Kobayashi M., Ashman K., Mitchell M., Rice G.E. Exosomal signaling during hypoxia mediates microvascular endothelial cell migration and vasculogenesis. PLoS One. 2013;8:e68451.
    1. Magatti M., de Munari S., Vertua E., Gibelli L., Wengler G.S., Parolini O. Human amnion mesenchyme harbors cells with allogeneic T-cell suppression and stimulation capabilities. Stem Cells. 2008;26:182–192. doi: 10.1634/stemcells.2007-0491.
    1. Wolbank S., Peterbauer A., Fahrner M., Hennerbichler S., van Griensven M., Stadler G., Redl H., Gabriel C. Dose-dependent immunomodulatory effect of human stem cells from amniotic membrane: A comparison with human mesenchymal stem cells from adipose tissue. Tissue Eng. 2007;13:1173–1183. doi: 10.1089/ten.2006.0313.
    1. Kim K.S., Kim H.S., Park J.M., Kim H.W., Park M.K., Lee H.S., Lim D.S., Lee T.H., Chopp M., Moon J. Long-term immunomodulatory effect of amniotic stem cells in an Alzheimer’s disease model. Neurobiol. Aging. 2013;34:2408–2420. doi: 10.1016/j.neurobiolaging.2013.03.029.
    1. Kim J.Y., Song S.H., Kim K.L., Ko J.J., Im J.E., Yie S.W., Ahn Y.K., Kim D.K., Suh W. Human cord blood-derived endothelial progenitor cells and their conditioned media exhibit therapeutic equivalence for diabetic wound healing. Cell Transplant. 2010;19:1635–1644. doi: 10.3727/096368910X516637.
    1. Park J.H., Hwang I., Hwang S.H., Han H., Ha H. Human umbilical cord blood-derived mesenchymal stem cells prevent diabetic renal injury through paracrine action. Diabetes Res. Clin. Pract. 2012;98:465–473. doi: 10.1016/j.diabres.2012.09.034.
    1. Wang H.S., Hung S.C., Peng S.T., Huang C.C., Wei H.M., Guo Y.J., Fu Y.S., Lai M.C., Chen C.C. Mesenchymal stem cells in the Wharton’s jelly of the human umbilical cord. Stem Cells. 2004;22:1330–1337. doi: 10.1634/stemcells.2004-0013.
    1. Yang C.C., Shih Y.H., Ko M.H., Hsu S.Y., Cheng H., Fu Y.S. Transplantation of human umbilical mesenchymal stem cells from Wharton’s jelly after complete transection of the rat spinal cord. PLoS One. 2008;3:e3336.
    1. Weiss M.L., Medicetty S., Bledsoe A.R., Rachakatla R.S., Choi M., Merchav S., Luo Y., Rao M.S., Velagaleti G., Troyer D. Human umbilical cord matrix stem cells: Preliminary characterization and effect of transplantation in a rodent model of Parkinson’s disease. Stem Cells. 2006;24:781–792. doi: 10.1634/stemcells.2005-0330.
    1. Ribeiro C.A., Fraga J.S., Graos M., Neves N.M., Reis R.L., Gimble J.M., Sousa N., Salgado A.J. The secretome of stem cells isolated from the adipose tissue and Wharton jelly acts differently on central nervous system derived cell populations. Stem Cell Res. Ther. 2012;3:18. doi: 10.1186/scrt109.
    1. Hsieh J.Y., Wang H.W., Chang S.J., Liao K.H., Lee I.H., Lin W.S., Wu C.H., Lin W.Y., Cheng S.M. Mesenchymal stem cells from human umbilical cord express preferentially secreted factors related to neuroprotection, neurogenesis, and angiogenesis. PLoS One. 2013;8:e72604. doi: 10.1371/journal.pone.0072604.
    1. Shohara R., Yamamoto A., Takikawa S., Iwase A., Hibi H., Kikkawa F., Ueda M. Mesenchymal stromal cells of human umbilical cord Wharton’s jelly accelerate wound healing by paracrine mechanisms. Cytotherapy. 2012;14:1171–1181. doi: 10.3109/14653249.2012.706705.
    1. Pierro M., Ionescu L., Montemurro T., Vadivel A., Weissmann G., Oudit G., Emery D., Bodiga S., Eaton F., Peault B., et al. Short-term, long-term and paracrine effect of human umbilical cord-derived stem cells in lung injury prevention and repair in experimental bronchopulmonary dysplasia. Thorax. 2013;68:475–484. doi: 10.1136/thoraxjnl-2012-202323.
    1. Zhou Y., Xu H., Xu W., Wang B., Wu H., Tao Y., Zhang B., Wang M., Mao F., Yan Y., et al. Exosomes released by human umbilical cord mesenchymal stem cells protect against cisplatin-induced renal oxidative stress and apoptosis in vivo and in vitro. Stem Cell Res. Ther. 2013;4:1–13. doi: 10.1186/scrt149.
    1. In’t Anker P.S., Scherjon S.A., van der Kleijburg K.C., Noort W.A., Claas F.H., Willemze R., Fibbe W.E., Kanhai H.H. Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood. 2003;102:1548–1549. doi: 10.1182/blood-2003-04-1291.
    1. Prusa A.R., Marton E., Rosner M., Bernaschek G., Hengstschlager M. Oct-4-expressing cells in human amniotic fluid: A new source for stem cell research? Hum. Reprod. 2003;18:1489–1493. doi: 10.1093/humrep/deg279.
    1. Kaviani A., Perry T.E., Dzakovic A., Jennings R.W., Ziegler M.M., Fauza D.O. The amniotic fluid as a source of cells for fetal tissue engineering. J. Pediatr. Surg. 2001;36:1662–1665. doi: 10.1053/jpsu.2001.27945.
    1. Kaviani A., Guleserian K., Perry T.E., Jennings R.W., Ziegler M.M., Fauza D.O. Fetal tissue engineering from amniotic fluid. J. Am. Coll. Surg. 2003;196:592–597. doi: 10.1016/S1072-7515(02)01834-3.
    1. Roubelakis M.G., Tsaknakis G., Pappa K.I., Anagnou N.P., Watt S.M. Spindle shaped human mesenchymal stem/stromal cells from amniotic fluid promote neovascularization. PLoS One. 2013;8:e54747.
    1. Roubelakis M.G., Trohatou O., Anagnou N.P. Amniotic fluid and amniotic membrane stem cells: Marker discovery. Stem Cells Int. 2012;2012 doi: 10.1155/2012/107836.
    1. Roubelakis M.G., Bitsika V., Zagoura D., Trohatou O., Pappa K.I., Makridakis M., Antsaklis A., Vlahou A., Anagnou N.P. In vitro and in vivo properties of distinct populations of amniotic fluid mesenchymal progenitor cells. J. Cell. Mol. Med. 2011;15:1896–1913. doi: 10.1111/j.1582-4934.2010.01180.x.
    1. De Coppi P., Bartsch G., Jr., Siddiqui M.M., Xu T., Santos C.C., Perin L., Mostoslavsky G., Serre A.C., Snyder E.Y., Yoo J.J., et al. Isolation of amniotic stem cell lines with potential for therapy. Nat. Biotechnol. 2007;25:100–106. doi: 10.1038/nbt1274.
    1. Moschidou D., Mukherjee S., Blundell M.P., Drews K., Jones G.N., Abdulrazzak H., Nowakowska B., Phoolchund A., Lay K., Ramasamy T.S., et al. Valproic acid confers functional pluripotency to human amniotic fluid stem cells in a transgene-free approach. Mol. Ther. 2012;20:1953–1967. doi: 10.1038/mt.2012.117.
    1. Yoon B.S., Moon J.H., Jun E.K., Kim J., Maeng I., Kim J.S., Lee J.H., Baik C.S., Kim A., Cho K.S., et al. Secretory profiles and wound healing effects of human amniotic fluid-derived mesenchymal stem cells. Stem Cells Dev. 2010;19:887–902. doi: 10.1089/scd.2009.0138.
    1. Mirabella T., Cilli M., Carlone S., Cancedda R., Gentili C. Amniotic liquid derived stem cells as reservoir of secreted angiogenic factors capable of stimulating neo-arteriogenesis in an ischemic model. Biomaterials. 2011;32:3689–3699. doi: 10.1016/j.biomaterials.2011.01.071.
    1. Mirabella T., Hartinger J., Lorandi C., Gentili C., van Griensven M., Cancedda R. Proangiogenic soluble factors from amniotic fluid stem cells mediate the recruitment of endothelial progenitors in a model of ischemic fasciocutaneous flap. Stem Cells Dev. 2012;21:2179–2188. doi: 10.1089/scd.2011.0639.
    1. Zagoura D.S., Roubelakis M.G., Bitsika V., Trohatou O., Pappa K.I., Kapelouzou A., Antsaklis A., Anagnou N.P. Therapeutic potential of a distinct population of human amniotic fluid mesenchymal stem cells and their secreted molecules in mice with acute hepatic failure. Gut. 2012;61:894–906. doi: 10.1136/gutjnl-2011-300908.
    1. Pederiva F., Ghionzoli M., Pierro A., de Coppi P., Tovar J.A. Amniotic fluid stem cells rescue both in vitro and in vivo growth, innervation, and motility in nitrofen-exposed hypoplastic rat lungs through paracrine effects. Cell Transplant. 2013;22:1683–1694. doi: 10.3727/096368912X657756.
    1. Grisafi D., Pozzobon M., Dedja A., Vanzo V., Tomanin R., Porzionato A., Macchi V., Salmaso R., Scarpa M., Cozzi E., et al. Human amniotic fluid stem cells protect rat lungs exposed to moderate hyperoxia. Pediatr. Pulmonol. 2013;48:1070–1080. doi: 10.1002/ppul.22791.
    1. Sedrakyan S., Da Sacco S., Milanesi A., Shiri L., Petrosyan A., Varimezova R., Warburton D., Lemley K.V., de Filippo R.E., Perin L. Injection of amniotic fluid stem cells delays progression of renal fibrosis. J. Am. Soc. Nephrol. 2012;23:661–673. doi: 10.1681/ASN.2011030243.
    1. Bollini S., Cheung K.K., Riegler J., Dong X., Smart N., Ghionzoli M., Loukogeorgakis S.P., Maghsoudlou P., Dube K.N., Riley P.R., et al. Amniotic fluid stem cells are cardioprotective following acute myocardial infarction. Stem Cells Dev. 2011;20:1985–1994. doi: 10.1089/scd.2010.0424.
    1. Zani A., Cananzi M., Fascetti-Leon F., Lauriti G., Smith V.V., Bollini S., Ghionzoli M., D’Arrigo A., Pozzobon M., Piccoli M., et al. Amniotic fluid stem cells improve survival and enhance repair of damaged intestine in necrotising enterocolitis via a COX-2 dependent mechanism. Gut. 2013 doi: 10.1136/gutjnl-2012-303735.
    1. Iglesias D.M., El-Kares R., Taranta A., Bellomo F., Emma F., Besouw M., Levtchenko E., Toelen J., van den Heuvel L., Chu L., et al. Stem cell microvesicles transfer cystinosin to human cystinotic cells and reduce cystine accumulation in vitro. PLoS One. 2012;7:e42840. doi: 10.1371/journal.pone.0042840.
    1. Fuchs E., Tumbar T., Guasch G. Socializing with the neighbors: Stem cells and their niche. Cell. 2004;116:769–778. doi: 10.1016/S0092-8674(04)00255-7.
    1. Da Silva Meirelles L., Caplan A.I., Nardi N.B. In search of the in vivo identity of mesenchymal stem cells. Stem Cells. 2008;26:2287–2299. doi: 10.1634/stemcells.2007-1122.
    1. Phinney D.G., Sensebe L. Mesenchymal stromal cells: Misconceptions and evolving concepts. Cytotherapy. 2013;15:140–145. doi: 10.1016/j.jcyt.2012.11.005.
    1. Dominici M., Le Blanc K., Mueller I., Slaper-Cortenbach I., Marini F., Krause D., Deans R., Keating A., Prockop D., Horwitz E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315–317.
    1. Prockop D.J., Oh J.Y. Mesenchymal stem/stromal cells (MSCs): Role as guardians of inflammation. Mol. Ther. 2012;20:14–20. doi: 10.1038/mt.2011.211.
    1. Kean T.J., Lin P., Caplan A.I., Dennis J.E. MSCs: Delivery routes and engraftment, cell-targeting strategies, and immune modulation. Stem Cells Int. 2013;2013 doi: 10.1155/2013/732742.
    1. Dai W., Hale S.L., Martin B.J., Kuang J.Q., Dow J.S., Wold L.E., Kloner R.A. Allogeneic mesenchymal stem cell transplantation in postinfarcted rat myocardium: Short- and long-term effects. Circulation. 2005;112:214–223. doi: 10.1161/CIRCULATIONAHA.104.527937.
    1. Gnecchi M., He H., Liang O.D., Melo L.G., Morello F., Mu H., Noiseux N., Zhang L., Pratt R.E., Ingwall J.S., et al. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat. Med. 2005;11:367–368. doi: 10.1038/nm0405-367.
    1. Togel F., Hu Z., Weiss K., Isaac J., Lange C., Westenfelder C. Administered mesenchymal stem cells protect against ischemic acute renal failure through differentiation-independent mechanisms. Am. J. Physiol. Renal Physiol. 2005;289:31–42. doi: 10.1152/ajprenal.00007.2005.
    1. Caplan A.I., Correa D. The MSC: An injury drugstore. Cell Stem Cell. 2011;9:11–15. doi: 10.1016/j.stem.2011.06.008.
    1. Tasso R., Gaetani M., Molino E., Cattaneo A., Monticone M., Bachi A., Cancedda R. The role of bFGF on the ability of MSC to activate endogenous regenerative mechanisms in an ectopic bone formation model. Biomaterials. 2012;33:2086–2096. doi: 10.1016/j.biomaterials.2011.11.043.
    1. Tasso R., Ulivi V., Reverberi D., Lo Sicco C., Descalzi F., Cancedda R. In vivo implanted bone marrow-derived mesenchymal stem cells trigger a cascade of cellular events leading to the formation of an ectopic bone regenerative niche. Stem Cells Dev. 2013;22:3178–3191. doi: 10.1089/scd.2013.0313.
    1. Nemeth K., Leelahavanichkul A., Yuen P.S., Mayer B., Parmelee A., Doi K., Robey P.G., Leelahavanichkul K., Koller B.H., Brown J.M., et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat. Med. 2009;15:42–49. doi: 10.1038/nm.1905.
    1. Roddy G.W., Oh J.Y., Lee R.H., Bartosh T.J., Ylostalo J., Coble K., Rosa R.H., Jr., Prockop D.J. Action at a distance: Systemically administered adult stem/progenitor cells (MSCs) reduce inflammatory damage to the cornea without engraftment and primarily by secretion of TNF-alpha stimulated gene/protein 6. Stem Cells. 2011;29:1572–1579. doi: 10.1002/stem.708.
    1. Gyorgy B., Szabo T.G., Pasztoi M., Pal Z., Misjak P., Aradi B., Laszlo V., Pallinger E., Pap E., Kittel A., et al. Membrane vesicles, current state-of-the-art: Emerging role of extracellular vesicles. Cell. Mol. Life Sci. 2011;68:2667–2688. doi: 10.1007/s00018-011-0689-3.
    1. Baglio S.R., Pegtel D.M., Baldini N. Mesenchymal stem cell secreted vesicles provide novel opportunities in (stem) cell-free therapy. Front. Physiol. 2012;3:1–11.
    1. Bruno S., Grange C., Deregibus M.C., Calogero R.A., Saviozzi S., Collino F., Morando L., Busca A., Falda M., Bussolati B., et al. Mesenchymal stem cell-derived microvesicles protect against acute tubular injury. J. Am. Soc. Nephrol. 2009;20:1053–1067. doi: 10.1681/ASN.2008070798.
    1. Gatti S., Bruno S., Deregibus M.C., Sordi A., Cantaluppi V., Tetta C., Camussi G. Microvesicles derived from human adult mesenchymal stem cells protect against ischaemia-reperfusion-induced acute and chronic kidney injury. Nephrol. Dial. Transplant. 2011;26:1474–1483. doi: 10.1093/ndt/gfr015.
    1. Timmers L., Lim S.K., Arslan F., Armstrong J.S., Hoefer I.E., Doevendans P.A., Piek J.J., El Oakley R.M., Choo A., Lee C.N., et al. Reduction of myocardial infarct size by human mesenchymal stem cell conditioned medium. Stem Cell Res. 2007;1:129–137.
    1. Arslan F., Lai R.C., Smeets M.B., Akeroyd L., Choo A., Aguor E.N., Timmers L., van Rijen H.V., Doevendans P.A., Pasterkamp G., et al. Mesenchymal stem cell-derived exosomes increase ATP levels, decrease oxidative stress and activate PI3K/Akt pathway to enhance myocardial viability and prevent adverse remodeling after myocardial ischemia/reperfusion injury. Stem Cell Res. 2013;10:301–312. doi: 10.1016/j.scr.2013.01.002.
    1. Xin H., Li Y., Cui Y., Yang J.J., Zhang Z.G., Chopp M. Systemic administration of exosomes released from mesenchymal stromal cells promote functional recovery and neurovascular plasticity after stroke in rats. J. Cereb. Blood Flow Metab. 2013;33:1711–1715. doi: 10.1038/jcbfm.2013.152.
    1. Li T., Yan Y., Wang B., Qian H., Zhang X., Shen L., Wang M., Zhou Y., Zhu W., Li W., et al. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate liver fibrosis. Stem Cells Dev. 2013;22:845–854. doi: 10.1089/scd.2012.0395.
    1. Maumus M., Jorgensen C., Noel D. Mesenchymal stem cells in regenerative medicine applied to rheumatic diseases: Role of secretome and exosomes. Biochimie. 2013;95:2229–2234. doi: 10.1016/j.biochi.2013.04.017.
    1. Aliotta J.M., Sanchez-Guijo F.M., Dooner G.J., Johnson K.W., Dooner M.S., Greer K.A., Greer D., Pimentel J., Kolankiewicz L.M., Puente N., et al. Alteration of marrow cell gene expression, protein production, and engraftment into lung by lung-derived microvesicles: A novel mechanism for phenotype modulation. Stem Cells. 2007;25:2245–2256. doi: 10.1634/stemcells.2007-0128.
    1. Mouquet F., Pfister O., Jain M., Oikonomopoulos A., Ngoy S., Summer R., Fine A., Liao R. Restoration of cardiac progenitor cells after myocardial infarction by self-proliferation and selective homing of bone marrow-derived stem cells. Circ. Res. 2005;97:1090–1092. doi: 10.1161/01.RES.0000194330.66545.f5.
    1. Nakanishi C., Yamagishi M., Yamahara K., Hagino I., Mori H., Sawa Y., Yagihara T., Kitamura S., Nagaya N. Activation of cardiac progenitor cells through paracrine effects of mesenchymal stem cells. Biochem. Biophys. Res. Commun. 2008;374:11–16. doi: 10.1016/j.bbrc.2008.06.074.
    1. Loffredo F.S., Steinhauser M.L., Gannon J., Lee R.T. Bone marrow-derived cell therapy stimulates endogenous cardiomyocyte progenitors and promotes cardiac repair. Cell Stem Cell. 2011;8:389–398. doi: 10.1016/j.stem.2011.02.002.
    1. Gnecchi M., Zhang Z., Ni A., Dzau V.J. Paracrine mechanisms in adult stem cell signaling and therapy. Circ. Res. 2008;103:1204–1219. doi: 10.1161/CIRCRESAHA.108.176826.
    1. Malliaras K., Zhang Y., Seinfeld J., Galang G., Tseliou E., Cheng K., Sun B., Aminzadeh M., Marban E. Cardiomyocyte proliferation and progenitor cell recruitment underlie therapeutic regeneration after myocardial infarction in the adult mouse heart. EMBO Mol. Med. 2013;5:191–209. doi: 10.1002/emmm.201201737.
    1. Manuguerra-Gagne R., Boulos P.R., Ammar A., Leblond F.A., Krosl G., Pichette V., Lesk M.R., Roy D.C. Transplantation of mesenchymal stem cells promotes tissue regeneration in a glaucoma model through laser-induced paracrine factor secretion and progenitor cell recruitment. Stem Cells. 2013;31:1136–1148. doi: 10.1002/stem.1364.
    1. Tasso R., Fais F., Reverberi D., Tortelli F., Cancedda R. The recruitment of two consecutive and different waves of host stem/progenitor cells during the development of tissue-engineered bone in a murine model. Biomaterials. 2010;31:2121–2129. doi: 10.1016/j.biomaterials.2009.11.064.
    1. Mirabella T., Poggi A., Scaranari M., Mogni M., Lituania M., Baldo C., Cancedda R., Gentili C. Recruitment of host’s progenitor cells to sites of human amniotic fluid stem cells implantation. Biomaterials. 2011;32:4218–4227. doi: 10.1016/j.biomaterials.2010.12.028.
    1. Vrijsen K.R., Sluijter J.P., Schuchardt M.W., van Balkom B.W., Noort W.A., Chamuleau S.A., Doevendans P.A. Cardiomyocyte progenitor cell-derived exosomes stimulate migration of endothelial cells. J. Cell. Mol. Med. 2010;14:1064–1070.
    1. Sahoo S., Klychko E., Thorne T., Misener S., Schultz K.M., Millay M., Ito A., Liu T., Kamide C., Agrawal H., et al. Exosomes from human CD34(+) stem cells mediate their proangiogenic paracrine activity. Circ. Res. 2011;109:724–728. doi: 10.1161/CIRCRESAHA.111.253286.
    1. Lai R.C., Yeo R.W., Tan K.H., Lim S.K. Exosomes for drug delivery—A novel application for the mesenchymal stem cell. Biotechnol. Adv. 2013;109:724–728.
    1. Lai R.C., Yeo R.W., Tan K.H., Lim S.K. Mesenchymal stem cell exosome ameliorates reperfusion injury through proteomic complementation. Regen. Med. 2013;8:197–209. doi: 10.2217/rme.13.4.
    1. Lai R.C., Arslan F., Lee M.M., Sze N.S., Choo A., Chen T.S., Salto-Tellez M., Timmers L., Lee C.N., El Oakley R.M., et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010;4:214–222. doi: 10.1016/j.scr.2009.12.003.
    1. Tetta C., Ghigo E., Silengo L., Deregibus M.C., Camussi G. Extracellular vesicles as an emerging mechanism of cell-to-cell communication. Endocrine. 2013;44:11–19. doi: 10.1007/s12020-012-9839-0.
    1. Valadi H., Ekstrom K., Bossios A., Sjostrand M., Lee J.J., Lotvall 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. doi: 10.1038/ncb1596.
    1. Chen T.S., Lai R.C., Lee M.M., Choo A.B., Lee C.N., Lim S.K. Mesenchymal stem cell secretes microparticles enriched in pre-microRNAs. Nucleic Acids Res. 2010;38:215–224. doi: 10.1093/nar/gkp857.
    1. Camussi G., Deregibus M.C., Tetta C. Paracrine/Endocrine mechanism of stem cells on kidney repair: Role of microvesicle-mediated transfer of genetic information. Curr. Opin. Nephrol. Hypertens. 2010;19:7–12. doi: 10.1097/MNH.0b013e328332fb6f.
    1. Cantaluppi V., Gatti S., Medica D., Figliolini F., Bruno S., Deregibus M.C., Sordi A., Biancone L., Tetta C., Camussi G. Microvesicles derived from endothelial progenitor cells protect the kidney from ischemia-reperfusion injury by microRNA-dependent reprogramming of resident renal cells. Kidney Int. 2012;82:412–427. doi: 10.1038/ki.2012.105.
    1. Ranghino A., Cantaluppi V., Grange C., Vitillo L., Fop F., Biancone L., Deregibus M.C., Tetta C., Segoloni G.P., Camussi G. Endothelial progenitor cell-derived microvesicles improve neovascularization in a murine model of hindlimb ischemia. Int. J. Immunopathol. Pharmacol. 2012;25:75–85.
    1. Xin H., Li Y., Buller B., Katakowski M., Zhang Y., Wang X., Shang X., Zhang Z.G., Chopp M. Exosome-mediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth. Stem Cells. 2012;30:1556–1564. doi: 10.1002/stem.1129.
    1. Yu B., Gong M., Wang Y., Millard R.W., Pasha Z., Yang Y., Ashraf M., Xu M. Cardiomyocyte protection by GATA-4 gene engineered mesenchymal stem cells is partially mediated by translocation of miR-221 in microvesicles. PLoS One. 2013;8:e73304.
    1. Ranganath S.H., Levy O., Inamdar M.S., Karp J.M. Harnessing the mesenchymal stem cell secretome for the treatment of cardiovascular disease. Cell Stem Cell. 2012;10:244–258. doi: 10.1016/j.stem.2012.02.005.
    1. Kim H.S., Choi D.Y., Yun S.J., Choi S.M., Kang J.W., Jung J.W., Hwang D., Kim K.P., Kim D.W. Proteomic analysis of microvesicles derived from human mesenchymal stem cells. J. Proteome Res. 2012;11:839–849. doi: 10.1021/pr200682z.
    1. Collino F., Deregibus M.C., Bruno S., Sterpone L., Aghemo G., Viltono L., Tetta C., Camussi G. Microvesicles derived from adult human bone marrow and tissue specific mesenchymal stem cells shuttle selected pattern of miRNAs. PLoS One. 2010;5:e11803. doi: 10.1371/journal.pone.0011803.
    1. Simpson R.J., Kalra H., Mathivanan S. ExoCarta as a resource for exosomal research. J. Extracell. Vesicles. 2012;1:1–6.
    1. Sanchez C., Oskowitz A., Pochampally R.R. Epigenetic reprogramming of IGF1 and leptin genes by serum deprivation in multipotential mesenchymal stromal cells. Stem Cells. 2009;27:375–382. doi: 10.1634/stemcells.2008-0546.
    1. Oskowitz A., McFerrin H., Gutschow M., Carter M.L., Pochampally R. Serum-deprived human multipotent mesenchymal stromal cells (MSCs) are highly angiogenic. Stem Cell Res. 2011;6:215–225. doi: 10.1016/j.scr.2011.01.004.
    1. Liu H., Liu S., Li Y., Wang X., Xue W., Ge G., Luo X. The role of SDF-1-CXCR4/CXCR7 axis in the therapeutic effects of hypoxia-preconditioned mesenchymal stem cells for renal ischemia/reperfusion injury. PLoS One. 2012;7:e34608.
    1. Tsai C.C., Yew T.L., Yang D.C., Huang W.H., Hung S.C. Benefits of hypoxic culture on bone marrow multipotent stromal cells. Am. J. Blood Res. 2012;2:148–159.
    1. Gnecchi M., He H., Noiseux N., Liang O.D., Zhang L., Morello F., Mu H., Melo L.G., Pratt R.E., Ingwall J.S., et al. Evidence supporting paracrine hypothesis for Akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvement. FASEB J. 2006;20:661–669. doi: 10.1096/fj.05-5211com.
    1. Haider H., Jiang S., Idris N.M., Ashraf M. IGF-1-Overexpressing mesenchymal stem cells accelerate bone marrow stem cell mobilization via paracrine activation of SDF-1alpha/CXCR4 signaling to promote myocardial repair. Circ. Res. 2008;103:1300–1308. doi: 10.1161/CIRCRESAHA.108.186742.
    1. Tang Y.L., Zhao Q., Qin X., Shen L., Cheng L., Ge J., Phillips M.I. Paracrine action enhances the effects of autologous mesenchymal stem cell transplantation on vascular regeneration in rat model of myocardial infarction. Ann. Thorac. Surg. 2005;80:229–236. doi: 10.1016/j.athoracsur.2005.02.072.
    1. Li H., Zuo S., He Z., Yang Y., Pasha Z., Wang Y., Xu M. Paracrine factors released by GATA-4 overexpressed mesenchymal stem cells increase angiogenesis and cell survival. Am. J. Physiol. Heart Circ. Physiol. 2010;299:1772–1781. doi: 10.1152/ajpheart.00557.2010.
    1. Yang F., Cho S.W., Son S.M., Bogatyrev S.R., Singh D., Green J.J., Mei Y., Park S., Bhang S.H., Kim B.S., et al. Genetic engineering of human stem cells for enhanced angiogenesis using biodegradable polymeric nanoparticles. Proc. Natl. Acad. Sci. USA. 2010;107:3317–3322. doi: 10.1073/pnas.0905432106.
    1. Yao Y., Zhang F., Wang L., Zhang G., Wang Z., Chen J., Gao X. Lipopolysaccharide preconditioning enhances the efficacy of mesenchymal stem cells transplantation in a rat model of acute myocardial infarction. J. Biomed. Sci. 2009;16 doi: 10.1186/1423-0127-16-74.
    1. Herrmann J.L., Wang Y., Abarbanell A.M., Weil B.R., Tan J., Meldrum D.R. Preconditioning mesenchymal stem cells with transforming growth factor-alpha improves mesenchymal stem cell-mediated cardioprotection. Shock. 2010;33:24–30. doi: 10.1097/SHK.0b013e3181b7d137.
    1. Lee M.J., Kim J., Kim M.Y., Bae Y.S., Ryu S.H., Lee T.G., Kim J.H. Proteomic analysis of tumor necrosis factor-alpha-induced secretome of human adipose tissue-derived mesenchymal stem cells. J. Proteome Res. 2010;9:1754–1762. doi: 10.1021/pr900898n.
    1. Croitoru-Lamoury J., Lamoury F.M., Zaunders J.J., Veas L.A., Brew B.J. Human mesenchymal stem cells constitutively express chemokines and chemokine receptors that can be upregulated by cytokines, IFN-beta, and copaxone. J. Interferon Cytokine Res. 2007;27:53–64. doi: 10.1089/jir.2006.0037.
    1. Block G.J., Ohkouchi S., Fung F., Frenkel J., Gregory C., Pochampally R., DiMattia G., Sullivan D.E., Prockop D.J. Multipotent stromal cells are activated to reduce apoptosis in part by upregulation and secretion of stanniocalcin-1. Stem Cells. 2009;27:670–681.
    1. Chen T.S., Arslan F., Yin Y., Tan S.S., Lai R.C., Choo A.B., Padmanabhan J., Lee C.N., de Kleijn D.P., Lim S.K. Enabling a robust scalable manufacturing process for therapeutic exosomes through oncogenic immortalization of human ESC-derived MSCs. J. Transl. Med. 2011;9:1–10. doi: 10.1186/1479-5876-9-1.
    1. Kupcova Skalnikova H. Proteomic techniques for characterisation of mesenchymal stem cell secretome. Biochimie. 2013;95:2196–2211. doi: 10.1016/j.biochi.2013.07.015.
    1. Ong S.E., Blagoev B., Kratchmarova I., Kristensen D.B., Steen H., Pandey A., Mann M. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol. Cell. Proteomics. 2002;1:376–386. doi: 10.1074/mcp.M200025-MCP200.
    1. Kang D., Oh S., Ahn S.M., Lee B.H., Moon M.H. Proteomic analysis of exosomes from human neural stem cells by flow field-flow fractionation and nanoflow liquid chromatography-tandem mass spectrometry. J. Proteome Res. 2008;7:3475–3480. doi: 10.1021/pr800225z.
    1. Lai R.C., Tan S.S., Teh B.J., Sze S.K., Arslan F., de Kleijn D.P., Choo A., Lim S.K. Proteolytic potential of the MSC exosome proteome: Implications for an exosome-mediated delivery of therapeutic proteasome. Int. J. Proteomics. 2012;2012 doi: 10.1155/2012/971907.
    1. Chen T.S., Lim S.K. Measurement of precursor miRNA in exosomes from human ESC-derived mesenchymal stem cells. Methods Mol. Biol. 2013;1024:69–86. doi: 10.1007/978-1-62703-453-1_6.
    1. Biancone L., Bruno S., Deregibus M.C., Tetta C., Camussi G. Therapeutic potential of mesenchymal stem cell-derived microvesicles. Nephrol. Dial. Transplant. 2012;27:3037–3042. doi: 10.1093/ndt/gfs168.

Source: PubMed

3
订阅