Human mesenchymal stem cells - current trends and future prospective

Imran Ullah, Raghavendra Baregundi Subbarao, Gyu Jin Rho, Imran Ullah, Raghavendra Baregundi Subbarao, Gyu Jin Rho

Abstract

Stem cells are cells specialized cell, capable of renewing themselves through cell division and can differentiate into multi-lineage cells. These cells are categorized as embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) and adult stem cells. Mesenchymal stem cells (MSCs) are adult stem cells which can be isolated from human and animal sources. Human MSCs (hMSCs) are the non-haematopoietic, multipotent stem cells with the capacity to differentiate into mesodermal lineage such as osteocytes, adipocytes and chondrocytes as well ectodermal (neurocytes) and endodermal lineages (hepatocytes). MSCs express cell surface markers like cluster of differentiation (CD)29, CD44, CD73, CD90, CD105 and lack the expression of CD14, CD34, CD45 and HLA (human leucocyte antigen)-DR. hMSCs for the first time were reported in the bone marrow and till now they have been isolated from various tissues, including adipose tissue, amniotic fluid, endometrium, dental tissues, umbilical cord and Wharton's jelly which harbours potential MSCs. hMSCs have been cultured long-term in specific media without any severe abnormalities. Furthermore, MSCs have immunomodulatory features, secrete cytokines and immune-receptors which regulate the microenvironment in the host tissue. Multilineage potential, immunomodulation and secretion of anti-inflammatory molecules makes MSCs an effective tool in the treatment of chronic diseases. In the present review, we have highlighted recent research findings in the area of hMSCs sources, expression of cell surface markers, long-term in vitro culturing, in vitro differentiation potential, immunomodulatory features, its homing capacity, banking and cryopreservation, its application in the treatment of chronic diseases and its use in clinical trials.

Figures

Figure 1. Number of clinical trials registered…
Figure 1. Number of clinical trials registered (per year) for MSCs based therapy (ClinicalTrials.gov)
Figure 2. Number of common diseases registered…
Figure 2. Number of common diseases registered for MSCs based cell therapy (ClinicalTrials.gov)

References

    1. Wei X., Yang X., Han Z.P., Qu F.F., Shao L., Shi Y.F. Mesenchymal stem cells: a new trend for cell therapy. Acta Pharmacol. Sin. 2013;34:747–754. doi: 10.1038/aps.2013.50.
    1. Evans M.J., Kaufman M.H. Establishment in culture of pluripotential cells from mouse embryos. Nature. 1981;292:154–156. doi: 10.1038/292154a0.
    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. Yoon S.W., Kim D.K., Kim K.P., Park K.S. Rad51 regulates cell cycle progression by preserving G2/M transition in mouse embryonic stem cells. Stem Cells Dev. 2014;23:2700–2711. doi: 10.1089/scd.2014.0129.
    1. Takahashi K., Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–676. doi: 10.1016/j.cell.2006.07.024.
    1. Takahashi K., Tanabe K., Ohnuki M., Narita M., Ichisaka T., Tomoda K., Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–872. doi: 10.1016/j.cell.2007.11.019.
    1. Nakagawa M., Koyanagi M., Tanabe K., Takahashi K., Ichisaka T., Aoi T., Okita K., Mochiduki Y., Takizawa N., Yamanaka S. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat. Biotechnol. 2008;26:101–106. doi: 10.1038/nbt1374.
    1. Friedenstein A.J., Gorskaja J.F., Kulagina N.N. Fibroblast precursors in normal and irradiated mouse hematopoietic organs. Exp. Hematol. 1976;4:267–274.
    1. Horwitz E.M., Le Blanc K., Dominici M., Mueller I., Slaper-Cortenbach I., Marini F.C., Deans R.J., Krause D.S., Keating A. International Society for Cellular Therapy. Clarification of the nomenclature for MSC: The International Society for Cellular Therapy position statement. Cytotherapy. 2005;7:393–395. doi: 10.1080/14653240500319234.
    1. Pittenger M.F., Mackay A.M., Beck S.C., Jaiswal R.K., Douglas R., Mosca J.D., Moorman M.A., Simonetti D.W., Craig S., Marshak D. R. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–147. doi: 10.1126/science.284.5411.143.
    1. Crisan M., Yap S., Casteilla L., Chen C.W., Corselli M., Park T.S., Andriolo G., Sun B., Zheng B., Zhang L., et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 2008;3:301–313. doi: 10.1016/j.stem.2008.07.003.
    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. doi: 10.1080/14653240600855905.
    1. Wagner W., Wein F., Seckinger A., Frankhauser M., Wirkner U., Krause U., Blake J., Schwager C., Eckstein V., Ansorge W., Ho A.D. Comparative characteristics of mesenchymal stem cells from human bone marrow, adipose tissue, and umbilical cord blood. Exp. Hematol. 2005;33:1402–1416. doi: 10.1016/j.exphem.2005.07.003.
    1. Zhang X., Yang M., Lin L., Chen P., Ma K.T., Zhou C.Y., Ao Y.F. Runx2 overexpression enhances osteoblastic differentiation and mineralization in adipose–derived stem cells in vitro and in vivo. Calcif. Tissue Int. 2006;79:169–178. doi: 10.1007/s00223-006-0083-6.
    1. In ’t Anker P.S., Scherjon S.A., Kleijburg-van der Keur 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. Tsai M.S., Lee J.L., Chang Y.J., Hwang S.M. Isolation of human multipotent mesenchymal stem cells from second-trimester amniotic fluid using a novel two-stage culture protocol. Hum. Reprod. 2004;19:1450–1456. doi: 10.1093/humrep/deh279.
    1. Cai J., Li W., Su H., Qin D., Yang J., Zhu F., Xu J., He W., Guo X., Labuda K., et al. Generation of human induced pluripotent stem cells from umbilical cord matrix and amniotic membrane mesenchymal cells. J. Biol. Chem. 2010;285:11227–11234. doi: 10.1074/jbc.M109.086389.
    1. Huang G.T., Gronthos S., Shi S. Mesenchymal stem cells derived from dental tissues vs. those from other sources: their biology and role in regenerative medicine. J. Dent. Res. 2009;88:792–806. doi: 10.1177/0022034509340867.
    1. Seifrtova M., Havelek R., Cmielova J., Jiroutova A., Soukup T., Bruckova L., Mokry J., English D., Rezacova M. The response of human ectomesenchymal dental pulp stem cells to cisplatin treatment. Int. Endod. J. 2012;45:401–412. doi: 10.1111/j.1365-2591.2011.01990.x.
    1. Schuring A.N., Schulte N., Kelsch R., Ropke A., Kiesel L., Gotte M. Characterization of endometrial mesenchymal stem-like cells obtained by endometrial biopsy during routine diagnostics. Fertil. Steril. 2011;95:423–426. doi: 10.1016/j.fertnstert.2010.08.035.
    1. Jiao F., Wang J., Dong Z.L., Wu M.J., Zhao T.B., Li D.D., Wang X. Human mesenchymal stem cells derived from limb bud can differentiate into all three embryonic germ layers lineages. Cell Reprogram. 2012;14:324–333.
    1. Allickson J.G., Sanchez A., Yefimenko N., Borlongan C.V., Sanberg P.R. Recent studies assessing the proliferative capability of a novel adult stem cell identified in menstrual blood. Open Stem Cell J. 2011;3:4–10. doi: 10.2174/1876893801103010004.
    1. Ab Kadir R., Zainal Ariffin S.H., Megat Abdul Wahab R., Kermani S., Senafi S. Characterization of mononucleated human peripheral blood cells. ScientificWorldJournal. 2012;2012:843843. doi: 10.1100/2012/843843.
    1. Raynaud C.M., Maleki M., Lis R., Ahmed B., Al-Azwani I., Malek J., Safadi F.F., Rafii A. Comprehensive characterization of mesenchymal stem cells from human placenta and fetal membrane and their response to osteoactivin stimulation. Stem Cells Int. 2012;2012:658356. doi: 10.1155/2012/658356.
    1. Rotter N., Oder J., Schlenke P., Lindner U., Bohrnsen F., Kramer J., Rohwedel J., Huss R., Brandau S., Wollenberg B., Lang S. Isolation and characterization of adult stem cells from human salivary glands. Stem Cells Dev. 2008;17:509–518. doi: 10.1089/scd.2007.0180.
    1. Bartsch G., Yoo J.J., De Coppi P., Siddiqui M.M., Schuch G., Pohl H.G., Fuhr J., Perin L., Soker S., Atala A. Propagation, expansion, and multilineage differentiation of human somatic stem cells from dermal progenitors. Stem Cells Dev. 2005;14:337–348. doi: 10.1089/scd.2005.14.337.
    1. Riekstina U., Muceniece R., Cakstina I., Muiznieks I., Ancans J. Characterization of human skin-derived mesenchymal stem cell proliferation rate in different growth conditions. Cytotechnology. 2008;58:153–162. doi: 10.1007/s10616-009-9183-2.
    1. Kita K., Gauglitz G.G., Phan T.T., Herndon D.N., Jeschke M.G. Isolation and characterization of mesenchymal stem cells from the sub-amniotic human umbilical cord lining membrane. Stem Cells Dev. 2010;19:491–502. doi: 10.1089/scd.2009.0192.
    1. Morito T., Muneta T., Hara K., Ju Y.J., Mochizuki T., Makino H., Umezawa A., Sekiya I. Synovial fluid-derived mesenchymal stem cells increase after intra-articular ligament injury in humans. Rheumatology. 2008;47:1137–1143. doi: 10.1093/rheumatology/ken114.
    1. Wang H.S., Hung S.C., Peng S.T., Chen C.C. Mesenchymal stem cells in the Wharton's jelly of the human umbilical cord. Stem Cells. 2004;22:330–1337. doi: 10.1634/stemcells.2004-0013.
    1. Hou T., Xu J., Wu X., Xie Z., Luo F., Zhang Z., Zeng L. Umbilical cord Wharton's Jelly: a new potential cell source of mesenchymal stromal cells for bone tissue engineering. Tissue Eng. Part A. 2009;15:2325–2334. doi: 10.1089/ten.tea.2008.0402.
    1. Kuznetsov S.A., Krebsbach P.H., Satomura K., Kerr J., Riminucci M., Benayahu D., Robey P.G. Single-colony derived strains of human marrow stromal fibroblasts form bone after transplantation in vivo. J. Bone Miner Res. 1997;12:1335–1347. doi: 10.1359/jbmr.1997.12.9.1335.
    1. Wu X.B., Tao R. Hepatocyte differentiation of mesenchymal stem cells. Hepatobiliary Pancreat. Dis. Int. 2012;11:360–371. doi: 10.1016/S1499-3872(12)60193-3.
    1. Pendleton C., Li Q., Chesler D.A., Yuan K., Guerrero-Cazares H., Quinones-Hinojosa A. Mesenchymal stem cells derived from adipose tissue vs bone marrow: in vitro comparison of their tropism towards gliomas. PLoS One. 2013;8:e58198. doi: 10.1371/journal.pone.0058198.
    1. Thirumala S., Goebel W.S., Woods E.J. Clinical grade adult stem cell banking. Organogenesis. 2009;5:143–154. doi: 10.4161/org.5.3.9811.
    1. Mamidi M.K., Nathan K.G., Singh G., Thrichelvam S.T., Mohd Yusof N.A., Fakharuzi N.A., Zakaria Z., Bhonde R., Das A.K., Majumdar A.S. Comparative cellular and molecular analyses of pooled bone marrow multipotent mesenchymal stromal cells during continuous passaging and after successive cryopreservation. J. Cell Biochem. 2012;113:3153–3164. doi: 10.1002/jcb.24193.
    1. Chamberlain G., Fox J., Ashton B., Middleton J. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells. 2007;25:2739–2749. doi: 10.1634/stemcells.2007-0197.
    1. Otsuru S., Hofmann T.J., Olson T.S., Dominici M., Horwitz E.M. Improved isolation and expansion of bone marrow mesenchymal stromal cells using a novel marrow filter device. Cytotherapy. 2013;15:146–153. doi: 10.1016/j.jcyt.2012.10.012.
    1. Tabatabaei M., Mosaffa N., Nikoo S., Bozorgmehr M., Ghods R., Kazemnejad S., Rezania S., Keshavarzi B., Arefi S., Ramezani-Tehrani F., et al. Isolation and partial characterization of human amniotic epithelial cells: the effect of trypsin. Avicenna J. Med. Biotechnol. 2014;6:10–20.
    1. Ranera B., Remacha A.R., Alvarez-Arguedas S., Castiella T., Vazquez F.J., Romero A., Zaragoza P., Martin-Burriel I., Rodellar C. Expansion under hypoxic conditions enhances the chondrogenic potential of equine bone marrow-derived mesenchymal stem cells. Vet. J. 2013;195:248–251. doi: 10.1016/j.tvjl.2012.06.008.
    1. Lin C.S., Ning H., Lin G., Lue T.F. Is CD34 truly a negative marker for mesenchymal stromal cells? Cytotherapy. 2012;14:1159–1163. doi: 10.3109/14653249.2012.729817.
    1. Vaculik C., Schuster C., Bauer W., Iram N., Pfisterer K., Kramer G., Reinisch A., Strunk D., Elbe-Burger A. Human dermis harbors distinct mesenchymal stromal cell subsets. J. Invest. Dermatol. 2012;132(3 Pt 1):563–574. doi: 10.1038/jid.2011.355.
    1. Zhang X., Hirai M., Cantero S., Ciubotariu R., Dobrila L., Hirsh A., Igura K., Satoh H., Yokomi I., Nishimura T., et al. Isolation and characterization of mesenchymal stem cells from human umbilical cord blood: reevaluation of critical factors for successful isolation and high ability to proliferate and differentiate to chondrocytes as compared to mesenchymal stem cells from bone marrow and adipose tissue. J. Cell. Biochem. 2011;112:1206–1218. doi: 10.1002/jcb.23042.
    1. Castrechini N.M., Murthi P., Qin S., Kusuma G.D., Wilton L., Abumaree M., Gronthos S., Zannettino A., Gude N.M., Brennecke S.P., Kalionis B. Decidua parietalis-derived mesenchymal stromal cells reside in a vascular niche within the choriodecidua. Reprod Sci. 2012;19:1302–1314. doi: 10.1177/1933719112450334.
    1. Park J.C., Kim J.M., Jung I.H., Kim J.C., Choi S.H., Cho K.S., Kim C.S. Isolation and characterization of human periodontal ligament (PDL) stem cells (PDLSCs) from the inflamed PDL tissue: in vitro and in vivo evaluations. J. Clin. Periodontol. 2011;38:721–731. doi: 10.1111/j.1600-051X.2011.01716.x.
    1. Kadar K., Kiraly M., Porcsalmy B., Molnar B., Racz G.Z., Blazsek J., Kallo K., Szabo E.L., Gera I., Gerber G., Varga G. Differentiation potential of stem cells from human dental origin - promise for tissue engineering. J. Physiol. Pharmacol. 2009;60(Suppl 7):167–175.
    1. Gronthos S., Graves S.E., Ohta S., Simmons P.J. The STRO-1+ fraction of adult human bone marrow contains the osteogenic precursors. Blood. 1994;84:4164–4173.
    1. Stewart K., Walsh S., Screen J., Jefferiss C.M., Chainey J., Jordan G.R., Beresford J.N. Further characterization of cells expressing STRO-1 in cultures of adult human bone marrow stromal cells. J. Bone Miner Res. 1999;14:1345–1356. doi: 10.1359/jbmr.1999.14.8.1345.
    1. Gronthos S., Franklin D.M., Leddy H.A., Robey P.G., Storms R.W., Gimble J.M. Surface protein characterization of human adipose tissue-derived stromal cells. J. Cell Physiol. 2001;189:54–63. doi: 10.1002/jcp.1138.
    1. Kassem M. Mesenchymal stem cells: biological characteristics and potential clinical applications. Cloning Stem Cells. 2004;6:369–374. doi: 10.1089/clo.2004.6.369.
    1. Bonab M.M., Alimoghaddam K., Talebian F., Ghaffari S.H., Ghavamzadeh A., Nikbin B. Aging of mesenchymal stem cell in vitro. BMC Cell Biol. 2006;7:14. doi: 10.1186/1471-2121-7-14.
    1. Colter D.C., Class R., DiGirolamo C.M., Prockop D.J. Rapid expansion of recycling stem cells in cultures of plastic-adherent cells from human bone marrow. Proc. Natl. Acad. Sci. U.S.A. 2000;97:3213–3218. doi: 10.1073/pnas.97.7.3213.
    1. Rosland G.V., Svendsen A., Torsvik A., Sobala E., McCormack E., Immervoll H., Mysliwietz J., Tonn J.C., Goldbrunner R., Lonning P.E., et al. Long-term cultures of bone marrow-derived human mesenchymal stem cells frequently undergo spontaneous malignant transformation. Cancer Res. 2009;69:5331–5339. doi: 10.1158/0008-5472.CAN-08-4630.
    1. Chen G., Yue A., Ruan Z., Yin Y., Wang R., Ren Y., Zhu L. Monitoring the biology stability of human umbilical cord-derived mesenchymal stem cells during long-term culture in serum-free medium. Cell Tissue Bank. 2014;15:513–521. doi: 10.1007/s10561-014-9420-6.
    1. Sensebe L. Clinical grade production of mesenchymal stem cells. Biomed. Mater Eng. 2008;18(1 Suppl):S3–S10.
    1. Majore I., Moretti P., Stahl F., Hass R., Kasper C. Growth and differentiation properties of mesenchymal stromal cell populations derived from whole human umbilical cord. Stem Cell Rev. 2011;7:17–31. doi: 10.1007/s12015-010-9165-y.
    1. Baglioni S., Francalanci M., Squecco R., Lombardi A., Cantini G., Angeli R., Gelmini S., Guasti D., Benvenuti S., Annunziato F., et al. Characterization of human adult stem-cell populations isolated from visceral and subcutaneous adipose tissue. FASEB J. 2009;23:3494–3505. doi: 10.1096/fj.08-126946.
    1. Moretti P., Hatlapatka T., Marten D., Lavrentieva A., Majore I., Hass R., Kasper C. Mesenchymal stromal cells derived from human umbilical cord tissues: primitive cells with potential for clinical and tissue engineering applications. Adv. Biochem. Eng. Biotechnol. 2010;123:29–54.
    1. Rosen E.D., MacDougald O.A. Adipocyte differentiation from the inside out. Nat. Rev. Mol. Cell Biol. 2006;7:885–896. doi: 10.1038/nrm2066.
    1. Muruganandan S., Roman A.A., Sinal C.J. Adipocyte differentiation of bone marrow-derived mesenchymal stem cells: cross talk with the osteoblastogenic program. Cell Mol. Life Sci. 2009;66:236–253. doi: 10.1007/s00018-008-8429-z.
    1. Neve A., Corrado A., Cantatore F.P. Osteoblast physiology in normal and pathological conditions. Cell Tissue Res. 2011;343:289–302. doi: 10.1007/s00441-010-1086-1.
    1. Pei L., Tontonoz P. Fat's loss is bone's gain. J. Clin. Invest. 2004;113:805–806. doi: 10.1172/JCI200421311.
    1. James A.W., Pang S., Askarinam A., Corselli M., Zara J.N., Goyal R., Chang L., Pan A., Shen J., Yuan W., et al. Additive effects of sonic hedgehog and Nell-1 signaling in osteogenic versus adipogenic differentiation of human adipose-derived stromal cells. Stem Cells Dev. 2012;21:2170–2178. doi: 10.1089/scd.2011.0461.
    1. Bennett C.N., Longo K.A., Wright W.S., Suva L.J., Lane T.F., Hankenson K.D., MacDougald O.A. Regulation of osteoblastogenesis and bone mass by Wnt10b. Proc. Natl. Acad. Sci. U.S.A. 2005;102:3324–3329. doi: 10.1073/pnas.0408742102.
    1. James A.W., Leucht P., Levi B., Carre A.L., Xu Y., Helms J.A., Longaker M.T. Sonic Hedgehog influences the balance of osteogenesis and adipogenesis in mouse adipose-derived stromal cells. Tissue Eng. Part A. 2010;16:2605–2616. doi: 10.1089/ten.tea.2010.0048.
    1. Fontaine C., Cousin W., Plaisant M., Dani C., Peraldi P. Hedgehog signaling alters adipocyte maturation of human mesenchymal stem cells. Stem Cells. 2008;26:1037–1046. doi: 10.1634/stemcells.2007-0974.
    1. Taipaleenmaki H., Abdallah B.M., AlDahmash A., Saamanen A.M., Kassem M. Wnt signalling mediates the cross-talk between bone marrow derived pre-adipocytic and pre-osteoblastic cell populations. Exp. Cell Res. 2011;317:745–756. doi: 10.1016/j.yexcr.2010.12.015.
    1. D'Alimonte I., Lannutti A., Pipino C., Di Tomo P., Pierdomenico L., Cianci E., Antonucci I., Marchisio M., Romano M., Stuppia L., et al. Wnt signaling behaves as a “master regulator” in the osteogenic and adipogenic commitment of human amniotic fluid mesenchymal stem cells. Stem Cell Rev. 2013;9:642–654. doi: 10.1007/s12015-013-9436-5.
    1. Kang Q., Song W.X., Luo Q., Tang N., Luo J., Luo X., Chen J., Bi Y., He B.C., Park J.K., et al. A comprehensive analysis of the dual roles of BMPs in regulating adipogenic and osteogenic differentiation of mesenchymal progenitor cells. Stem Cells Dev. 2009;18:545–559. doi: 10.1089/scd.2008.0130.
    1. Dorman L.J., Tucci M., Benghuzzi H. In vitro effects of bmp-2, bmp-7, and bmp-13 on proliferation and differentation of mouse mesenchymal stem cells. Biomed. Sci. Instrum. 2012;48:81–87.
    1. Chen D., Ji X., Harris M.A., Feng J.Q., Karsenty G., Celeste A.J., Rosen V., Mundy G.R., Harris S.E. Differential roles for bone morphogenetic protein (BMP) receptor type IB and IA in differentiation and specification of mesenchymal precursor cells to osteoblast and adipocyte lineages. J. Cell Biol. 1998;142:295–305. doi: 10.1083/jcb.142.1.295.
    1. Valenti M.T., Garbin U., Pasini A., Zanatta M., Stranieri C., Manfro S., Zucal C., Dalle Carbonare L. Role of ox-PAPCs in the differentiation of mesenchymal stem cells (MSCs) and Runx2 and PPARgamma2 expression in MSCs-like of osteoporotic patients. PLoS One. 2011;6:e20363. doi: 10.1371/journal.pone.0020363.
    1. Mackay A.M., Beck S.C., Murphy J.M., Barry F.P., Chichester C.O., Pittenger M.F. Chondrogenic differentiation of cultured human mesenchymal stem cells from marrow. Tissue Eng. 1998;4:415–428. doi: 10.1089/ten.1998.4.415.
    1. Archer W., Francis-West P. The chondrocyte. Int. J. Biochem. Cell Biol. 2003;35:401–404. doi: 10.1016/S1357-2725(02)00301-1.
    1. Chimal-Monroy J., Diaz de Leon L. Expression of N-cadherin, N-CAM, fibronectin and tenascin is stimulated by TGF-beta1, beta2, beta3 and beta5 during the formation of precartilage condensations. Int. J. Dev. Biol. 1999;43:59–67.
    1. Akiyama H., Chaboissier M.C., Martin J.F., Schedl A., de Crombrugghe B. The transcription factor Sox9 has essential roles in successive steps of the chondrocyte differentiation pathway and is required for expression of Sox5 and Sox6. Genes Dev. 2002;16:2813–2828. doi: 10.1101/gad.1017802.
    1. Ikeda T., Kamekura S., Mabuchi A., Kou I., Seki S., Takato T., Nakamura K., Kawaguchi H., Ikegawa S., Chung U.I. The combination of SOX5, SOX6, and SOX9 (the SOX trio) provides signals sufficient for induction of permanent cartilage. Arthritis Rheum. 2004;50:3561–3573. doi: 10.1002/art.20611.
    1. An C., Cheng Y., Yuan Q., Li J. IGF-1 and BMP-2 induces differentiation of adipose-derived mesenchymal stem cells into chondrocytes-like cells. Ann. Biomed. Eng. 2010;38:1647–1654. doi: 10.1007/s10439-009-9892-x.
    1. Zhou S. TGF-beta regulates beta-catenin signaling and osteoblast differentiation in human mesenchymal stem cells. J. Cell Biochem. 2011;112:1651–1660. doi: 10.1002/jcb.23079.
    1. Wei Y., Hu Y., Lv R., Li D. Regulation of adipose-derived adult stem cells differentiating into chondrocytes with the use of rhBMP-2. Cytotherapy. 2006;8:570–579. doi: 10.1080/14653240600987690.
    1. Lanske B., Kronenberg H.M. Parathyroid hormone-related peptide (PTHrP) and parathyroid hormone (PTH)/PTHrP receptor. Crit. Rev. Eukaryot. Gene Expr. 1998;8:297–320. doi: 10.1615/CritRevEukarGeneExpr.v8.i3-4.40.
    1. Weisser J., Riemer S., Schmidl M., Suva L.J., Poschl E., Brauer R., von der Mark K. Four distinct chondrocyte populations in the fetal bovine growth plate: highest expression levels of PTH/PTHrP receptor, Indian hedgehog, and MMP-13 in hypertrophic chondrocytes and their suppression by PTH (1–34) and PTHrP (1–40) Exp. Cell Res. 2002;279:1–13. doi: 10.1006/excr.2002.5580.
    1. Wakitani S., Saito T., Caplan A.I. Myogenic cells derived from rat bone marrow mesenchymal stem cells exposed to 5-azacytidine. Muscle Nerve. 1995;18:1417–1426. doi: 10.1002/mus.880181212.
    1. Xu W., Zhang X., Qian H., Zhu W., Sun X., Hu J., Zhou H., Chen Y. Mesenchymal stem cells from adult human bone marrow differentiate into a cardiomyocyte phenotype in vitro. Exp. Biol. Med. 2004;229:623–631.
    1. Dezawa M., Ishikawa H., Itokazu Y., Yoshihara T., Hoshino M., Takeda S., Ide C., Nabeshima Y. Bone marrow stromal cells generate muscle cells and repair muscle degeneration. Science. 2005;309:314–317. doi: 10.1126/science.1110364.
    1. Narita Y., Yamawaki A., Kagami H., Ueda M., Ueda Y. Effects of transforming growth factor-beta 1 and ascorbic acid on differentiation of human bone-marrow-derived mesenchymal stem cells into smooth muscle cell lineage. Cell Tissue Res. 2008;333:449–459. doi: 10.1007/s00441-008-0654-0.
    1. Naghdi M., Tiraihi T., Namin S.A., Arabkheradmand J. Transdifferentiation of bone marrow stromal cells into cholinergic neuronal phenotype: a potential source for cell therapy in spinal cord injury. Cytotherapy. 2009;11:137–152. doi: 10.1080/14653240802716582.
    1. Datta I., Mishra S., Mohanty L., Pulikkot S., Joshi P.G. Neuronal plasticity of human Wharton's jelly mesenchymal stromal cells to the dopaminergic cell type compared with human bone marrow mesenchymal stromal cells. Cytotherapy. 2011;13:918–932. doi: 10.3109/14653249.2011.579957.
    1. Barzilay R., Ben-Zur T., Bulvik S., Melamed E., Offen D. Lentiviral delivery of LMX1a enhances dopaminergic phenotype in differentiated human bone marrow mesenchymal stem cells. Stem. Cells Dev. 2009;18:591–601. doi: 10.1089/scd.2008.0138.
    1. Safford K.M., Hicok K.C., Safford S.D., Halvorsen Y.D., Wilkison W.O., Gimble J.M., Rice H.E. Neurogenic differentiation of murine and human adipose-derived stromal cells. Biochem. Biophys. Res. Commun. 2002;294:371–379. doi: 10.1016/S0006-291X(02)00469-2.
    1. Kang S.K., Lee D.H., Bae Y.C., Kim H.K., Baik S.Y., Jung J.S. Improvement of neurological deficits by intracerebral transplantation of human adipose tissue-derived stromal cells after cerebral ischemia in rats. Exp. Neurol. 2003;183:355–366. doi: 10.1016/S0014-4886(03)00089-X.
    1. Rooney G.E., Howard L., O'Brien T., Windebank A.J., Barry F.P. Elevation of cAMP in mesenchymal stem cells transiently upregulates neural markers rather than inducing neural differentiation. Stem Cells Dev. 2009;18:387–398. doi: 10.1089/scd.2008.0080.
    1. Anghileri E., Marconi S., Pignatelli A., Cifelli P., Galie M., Sbarbati A., Krampera M., Belluzzi O., Bonetti B. Neuronal differentiation potential of human adipose-derived mesenchymal stem cells. Stem Cells Dev. 2008;17:909–916. doi: 10.1089/scd.2007.0197.
    1. Schaffler A., Buchler C. Concise review: adipose tissue-derived stromal cells–basic and clinical implications for novel cell-based therapies. Stem Cells. 2007;25:818–827. doi: 10.1634/stemcells.2006-0589.
    1. Pavlova G., Lopatina T., Kalinina N., Rybalkina E., Parfyonova Y., Tkachuk V., Revishchin A. In vitro neuronal induction of adipose-derived stem cells and their fate after transplantation into injured mouse brain. Curr. Med. Chem. 2012;19:5170–5177. doi: 10.2174/092986712803530557.
    1. Moussavou G., Kwak D.H., Lim M.U., Kim J.S., Kim S.U., Chang K.T., Choo Y.K. Role of gangliosides in the differentiation of human mesenchymal-derived stem cells into osteoblasts and neuronal cells. BMB Rep. 2013;46:527–532. doi: 10.5483/BMBRep.2013.46.11.179.
    1. Zhao F., Qu Y., Liu H., Du B., Mu D. Umbilical cord blood mesenchymal stem cells co-modified by TERT and BDNF: A novel neuroprotective therapy for neonatal hypoxic-ischemic brain damage. Int. J. Dev. Neurosci. 2014;38C:147–154. doi: 10.1016/j.ijdevneu.2014.06.014.
    1. Vollner F., Ernst W., Driemel O., Morsczeck C. A two-step strategy for neuronal differentiation in vitro of human dental follicle cells. Differentiation. 2009;77:433–441. doi: 10.1016/j.diff.2009.03.002.
    1. Wang J., Wang X., Sun Z., Wang X., Yang H., Shi S., Wang S. Stem cells from human-exfoliated deciduous teeth can differentiate into dopaminergic neuron-like cells. Stem Cells Dev. 2010;19:1375–1383. doi: 10.1089/scd.2009.0258.
    1. Lee K.D., Kuo T.K., Whang-Peng J., Chung Y.F., Lin C.T., Chou S.H., Chen J.R., Chen Y.P., Lee O.K. In vitro hepatic differentiation of human mesenchymal stem cells. Hepatology. 2004;40:1275–1284. doi: 10.1002/hep.20469.
    1. Stock P., Bruckner S., Winkler S., Dollinger M.M., Christ B. Human bone marrow mesenchymal stem cell-derived hepatocytes improve the mouse liver after acute acetaminophen intoxication by preventing progress of injury. Int. J. Mol. Sci. 2014;15:7004–7028. doi: 10.3390/ijms15047004.
    1. Parviz F., Matullo C., Garrison W.D., Savatski L., Adamson J.W., Ning G., Kaestner K.H., Rossi J.M., Zaret K.S., Duncan S.A. Hepatocyte nuclear factor 4alpha controls the development of a hepatic epithelium and liver morphogenesis. Nat. Genet. 2003;34:292–296. doi: 10.1038/ng1175.
    1. Watt A.J., Garrison W.D., Duncan S.A. HNF4: a central regulator of hepatocyte differentiation and function. Hepatology. 2003;37:1249–1253. doi: 10.1053/jhep.2003.50273.
    1. Hang H., Yu Y., Wu N., Huang Q., Xia Q., Bian J. Induction of highly functional hepatocytes from human umbilical cord mesenchymal stem cells by HNF4alpha transduction. PLoS One. 2014;9:e104133. doi: 10.1371/journal.pone.0104133.
    1. An S.Y., Han J., Lim H.J., Park S.Y., Kim J.H., Do B.R., Kim J.H. Valproic acid promotes differentiation of hepatocyte-like cells from whole human umbilical cord-derived mesenchymal stem cells. Tissue Cell. 2014;46:127–135. doi: 10.1016/j.tice.2013.12.006.
    1. Tang D.Q., Wang Q., Burkhardt B.R., Litherland S.A., Atkinson M.A., Yang L.J. In vitro generation of functional insulin-producing cells from human bone marrow-derived stem cells, but long-term culture running risk of malignant transformation. Am. J. Stem. Cells. 2012;1:114–127.
    1. Gabr M.M., Zakaria M.M., Refaie A.F., Ismail A.M., Abou-El-Mahasen M.A., Ashamallah S.A., Khater S.M., El-Halawani S.M., Ibrahim R.Y., Uin G.S., et al. Insulin-producing cells from adult human bone marrow mesenchymal stem cells control streptozotocin-induced diabetes in nude mice. Cell Transplant. 2013;22:133–145. doi: 10.3727/096368912X647162.
    1. Phadnis S.M., Joglekar M.V., Dalvi M.P., Muthyala S., Nair P.D., Ghaskadbi S.M., Bhonde R.R., Hardikar A.A. Human bone marrow-derived mesenchymal cells differentiate and mature into endocrine pancreatic lineage in vivo. Cytotherapy. 2011;13:279–293. doi: 10.3109/14653249.2010.523108.
    1. Govindasamy V., Ronald V.S., Abdullah A.N., Nathan K.R., Ab Aziz Z.A., Abdullah M., Musa S., Kasim N.H., Bhonde R.R. Differentiation of dental pulp stem cells into islet-like aggregates. J. Dent. Res. 2011;90:646–652. doi: 10.1177/0022034510396879.
    1. Kim J., Park S., Kang H.M., Ahn C.W., Kwon H.C., Song J.H., Lee Y.J., Lee K.H., Yang H., Baek S.Y., et al. Human insulin secreted from insulinogenic xenograft restores normoglycemia in type 1 diabetic mice without immunosuppression. Cell Transplant. 2012;21:2131–2147. doi: 10.3727/096368912X636803.
    1. Criscimanna A., Zito G., Taddeo A., Richiusa P., Pitrone M., Morreale D., Lodato G., Pizzolanti G., Citarrella R., Galluzzo A., Giordano C. In vitro generation of pancreatic endocrine cells from human adult fibroblast-like limbal stem cells. Cell Transplant. 2012;21:73–90. doi: 10.3727/096368911X580635.
    1. Kanafi M.M., Rajeshwari Y.B., Gupta S., Dadheech N., Nair P.D., Gupta P.K., Bhonde R.R. Transplantation of islet-like cell clusters derived from human dental pulp stem cells restores normoglycemia in diabetic mice. Cytotherapy. 2013;15:1228–1236. doi: 10.1016/j.jcyt.2013.05.008.
    1. Rasmusson I., Ringden O., Sundberg B., Le Blanc K. Mesenchymal stem cells inhibit the formation of cytotoxic T lymphocytes, but not activated cytotoxic T lymphocytes or natural killer cells. Transplantation. 2003;76:1208–1213. doi: 10.1097/01.TP.0000082540.43730.80.
    1. Figueroa F.F., Carrión F., Villanueva S., Khoury M. Mesenchymal Stem Cell treatment for autoimmune diseases: a critical review. Biol. Res. 2012;45:269–77. doi: 10.4067/S0716-97602012000300008.
    1. Selmani Z., Naji A., Zidi I., Favier B., Gaiffe E., Obert L., Borg C., Saas P., Tiberghien P., Rouas-Freiss N., et al. Human leukocyte antigen-G5 secretion by human mesenchymal stem cells is required to suppress T lymphocyte and natural killer function and to induce CD4+CD25highFOXP3+ regulatory T cells. Stem Cells. 2008;26:212–222. doi: 10.1634/stemcells.2007-0554.
    1. Zhao Z.G., Li W.M., Chen Z.C., You Y., Zou P. Immunosuppressive properties of mesenchymal stem cells derived from bone marrow of patients with chronic myeloid leukemia. Immunol. Invest. 2008;37:726–739. doi: 10.1080/08820130802349940.
    1. Nasef A., Mazurier C., Bouchet S., Francois S., Chapel A., Thierry D., Gorin N.C., Fouillard L. Leukemia inhibitory factor: role in human mesenchymal stem cells mediated immunosuppression. Cell Immunol. 2008;253:16–22. doi: 10.1016/j.cellimm.2008.06.002.
    1. Sheng H., Wang Y., Jin Y., Zhang Q., Zhang Y., Wang L., Shen B., Yin S., Liu W., Cui L., Li N. A critical role of IFNgamma in priming MSC-mediated suppression of T cell proliferation through up-regulation of B7-H1. Cell Res. 2008;18:846–857. doi: 10.1038/cr.2008.80.
    1. Spaggiari G.M., Capobianco A., Becchetti S., Mingari M.C., Moretta L. Mesenchymal stem cell-natural killer cell interaction: evidence that activated NK cells are capable of killing MSCs, whereas MSCs can inhibit IL-2-induced NK-cell proliferation. Blood. 2006;107:1484–1490. doi: 10.1182/blood-2005-07-2775.
    1. Sundin M., Ringden O., Sundberg B., Nava S., Gotherstrom C., Le Blanc K. No alloantibodies against mesenchymal stromal cells, but presence of anti-fetal calf serum antibodies, after transplantation in allogeneic hematopoietic stem cell recipients. Haematologica. 2007;92:1208–1215. doi: 10.3324/haematol.11446.
    1. Stagg J. Immune regulation by mesenchymal stem cells: two sides to the coin. Tissue Antigens. 2007;69:1–9. doi: 10.1111/j.1399-0039.2006.00739.x.
    1. Jewett A., Arasteh A., Tseng H.C., Behel A., Arasteh H., Yang W., Cacalano N.A., Paranjpe A. Strategies to rescue mesenchymal stem cells (MSCs) and dental pulp stem cells (DPSCs) from NK cell mediated cytotoxicity. PLoS One. 2010;5:e9874. doi: 10.1371/journal.pone.0009874.
    1. Giuliani M., Bennaceur-Griscelli A., Nanbakhsh A., Oudrhiri N., Chouaib S., Azzarone B., Durrbach A., Lataillade J.J. TLR ligands stimulation protects MSC from NK killing. Stem Cells. 2014;32:290–300. doi: 10.1002/stem.1563.
    1. Mazzini L., Fagioli F., Boccaletti R., Mareschi K., Oliveri G., Olivieri C., Pastore I., Marasso R., Madon E. Stem cell therapy in amyotrophic lateral sclerosis: a methodological approach in humans. Amyotroph. Lateral Scler. Other Motor Neuron. Disord. 2003;4:158–161. doi: 10.1080/14660820310014653.
    1. Suzuki M., McHugh J., Tork C., Shelley B., Hayes A., Bellantuono I., Aebischer P., Svendsen C.N. Direct muscle delivery of GDNF with human mesenchymal stem cells improves motor neuron survival and function in a rat model of familial ALS. Mol. Ther. 2008;16:2002–2010. doi: 10.1038/mt.2008.197.
    1. Kan I., Ben-Zur T., Barhum Y., Levy Y.S., Burstein A., Charlow T., Bulvik S., Melamed E., Offen D. Dopaminergic differentiation of human mesenchymal stem cells–utilization of bioassay for tyrosine hydroxylase expression. Neurosci. Lett. 2007;419:28–33. doi: 10.1016/j.neulet.2007.03.070.
    1. Wang F., Yasuhara T., Shingo T., Kameda M., Tajiri N., Yuan W.J., Kondo A., Kadota T., Baba T., Tayra J.T., et al. Intravenous administration of mesenchymal stem cells exerts therapeutic effects on parkinsonian model of rats: focusing on neuroprotective effects of stromal cell-derived factor-1alpha. BMC Neurosci. 2010;11:52. doi: 10.1186/1471-2202-11-52.
    1. Wilkins A., Kemp K., Ginty M., Hares K., Mallam E., Scolding N. Human bone marrow-derived mesenchymal stem cells secrete brain-derived neurotrophic factor which promotes neuronal survival in vitro. Stem Cell Res. 2009;3:63–70. doi: 10.1016/j.scr.2009.02.006.
    1. Danielyan L., Beer-Hammer S., Stolzing A., Schafer R., Siegel G., Fabian C., Kahle P., Biedermann T., Lourhmati A., Buadze M., et al. Intranasal delivery of bone marrow-derived mesenchymal stem cells, macrophages, and microglia to the brain in mouse models of Alzheimer's and Parkinson's disease. Cell Transplant. 2014;23(Suppl 1):123–139. doi: 10.3727/096368914X684970.
    1. Borlongan C.V. Recent preclinical evidence advancing cell therapy for Alzheimer's disease. Exp. Neurol. 2012;237:142–146. doi: 10.1016/j.expneurol.2012.06.024.
    1. Ma T., Gong K., Ao Q., Yan Y., Song B., Huang H., Zhang X., Gong Y. Intracerebral transplantation of adipose-derived mesenchymal stem cells alternatively activates microglia and ameliorates neuropathological deficits in Alzheimer's disease mice. Cell Transplant. 2013;22(Suppl 1):S113–S126. doi: 10.3727/096368913X672181.
    1. Saresella M., Calabrese E., Marventano I., Piancone F., Gatti A., Calvo M.G., Nemni R., Clerici M. PD1 negative and PD1 positive CD4+ T regulatory cells in mild cognitive impairment and Alzheimer's disease. J. Alzheimers Dis. 2010;21:927–938.
    1. Walsh J.T., Kipnis J. Regulatory T cells in CNS injury: the simple, the complex and the confused. Trends Mol. Med. 2011;17:541–547. doi: 10.1016/j.molmed.2011.05.012.
    1. Yang H., Yang H., Xie Z., Wei L., Bi J. Systemic transplantation of human umbilical cord derived mesenchymal stem cells-educated T regulatory cells improved the impaired cognition in AbetaPPswe/PS1dE9 transgenic mice. PLoS One. 2013;8:e69129. doi: 10.1371/journal.pone.0069129.
    1. Shin J.Y., Park H.J., Kim H.N., Oh S.H., Bae J.S., Ha H.J., Lee P.H. Mesenchymal stem cells enhance autophagy and increase beta-amyloid clearance in Alzheimer disease models. Autophagy. 2014;10:32–44. doi: 10.4161/auto.26508.
    1. Riordan H., Chan K., Marleau A.M., Ichim T.E. Cord blood in regenerative medicine: do we need immune suppression? J. Transl. Med. 2007;5:8. doi: 10.1186/1479-5876-5-8.
    1. Gonzalez M.A., Gonzalez-Rey E., Rico L., Buscher D., Delgad M. Treatment of experimental arthritis by inducing immune tolerance with human adipose-derived mesenchymal stem cells. Arthritis Rheum. 2009;60:1006–1019. doi: 10.1002/art.24405.
    1. Wehrens E.J., Prakken B.J., van Wijk F. T cells out of control–impaired immune regulation in the inflamed joint. Nat. Rev. Rheumatol. 2013;9:34–42. doi: 10.1038/nrrheum.2012.149.
    1. Augello A., Tasso R., Negrini S.M., Cancedda R., Pennesi G. Cell therapy using allogeneic bone marrow mesenchymal stem cells prevents tissue damage in collagen-induced arthritis. Arthritis Rheum. 2007;56:1175–1186. doi: 10.1002/art.22511.
    1. Papadopoulou A., Yiangou M., Athanasiou E., Zogas N., Kaloyannidis P., Batsis I., Fassas A., Anagnostopoulos A., Yannaki E. Mesenchymal stem cells are conditionally therapeutic in preclinical models of rheumatoid arthritis. Ann. Rheum. Dis. 2012;71:1733–1740. doi: 10.1136/annrheumdis-2011-200985.
    1. Zulewski H., Abraham E.J., Gerlach M.J., Daniel P.B., Moritz W., Muller B., Vallejo M., Thomas M.K., Habener J.F. Multipotential nestin-positive stem cells isolated from adult pancreatic islets differentiate ex vivo into pancreatic endocrine, exocrine, and hepatic phenotypes. Diabetes. 2001;50:521–533. doi: 10.2337/diabetes.50.3.521.
    1. Huang H., Tang X. Phenotypic determination and characterization of nestin-positive precursors derived from human fetal pancreas. Lab. Invest. 2003;83:539–547. doi: 10.1097/01.LAB.0000062890.40534.1C.
    1. Prabakar K.R., Dominguez-Bendala J., Molano R.D., Pileggi A., Villate S., Ricordi C., Inverardi L. Generation of glucose-responsive, insulin-producing cells from human umbilical cord blood-derived mesenchymal stem cells. Cell Transplant. 2012;21:1321–1339. doi: 10.3727/096368911X612530.
    1. Unsal I.O., Ginis Z., Pinarli F.A., Albayrak A., Cakal E., Sahin M., Delibasi T. Comparison of therapeutic characteristics of islet cell transplantation simultaneous with pancreatic mesenchymal stem cell transplantation in rats with type 1 diabetes mellitus. Stem Cell Rev. 2014 in the press.
    1. Noort W.A., Feye D., Van Den Akker F., Stecher D., Chamuleau S.A., Sluijter J.P., Doevendans P.A. Mesenchymal stromal cells to treat cardiovascular disease: strategies to improve survival and therapeutic results. Panminerva Med. 2010;52:27–40.
    1. Ramkisoensing A.A., Pijnappels D.A., Askar S.F., Passier R., Swildens J., Goumans M.J., Schutte C.I., de Vries A.A., Scherjon S., Mummery C.L., et al. Human embryonic and fetal mesenchymal stem cells differentiate toward three different cardiac lineages in contrast to their adult counterparts. PLoS One. 2011;6:e24164. doi: 10.1371/journal.pone.0024164.
    1. Liu J., Hu Q., Wang Z., Xu C., Wang X., Gong G., Mansoor A., Lee J., Hou M., Zeng L., et al. Autologous stem cell transplantation for myocardial repair. Am. J. Physiol. Heart Circ. Physiol. 2004;287:H501–H511. doi: 10.1152/ajpheart.00019.2004.
    1. Shake J.G., Gruber P.J., Baumgartner W.A., Senechal G., Meyers J., Redmond J.M., Pittenger M.F., Martin B.J. Mesenchymal stem cell implantation in a swine myocardial infarct model: engraftment and functional effects. Ann. Thorac. Surg. 2002;73:1919–1925. doi: 10.1016/S0003-4975(02)03517-8. discussion 1926.
    1. Nagaya N., Kangawa K., Itoh T., Iwase T., Murakami S., Miyahara Y., Fujii T., Uematsu M., Ohgushi H., Yamagishi M., et al. Transplantation of mesenchymal stem cells improves cardiac function in a rat model of dilated cardiomyopathy. Circulation. 2005;112:1128–1135. doi: 10.1161/CIRCULATIONAHA.104.500447.
    1. Katritsis D.G., Sotiropoulou P.A., Karvouni E., Karabinos I., Korovesis S., Perez S.A., Voridis E.M., Papamichail M. Transcoronary transplantation of autologous mesenchymal stem cells and endothelial progenitors into infarcted human myocardium. Catheter Cardiovasc. Interv. 2005;65:321–329. doi: 10.1002/ccd.20406.
    1. Freyman T., Polin G., Osman H., Crary J., Lu M., Cheng L., Palasis M., Wilensky R.L. A quantitative, randomized study evaluating three methods of mesenchymal stem cell delivery following myocardial infarction. Eur. Heart J. 2006;27:1114–1122. doi: 10.1093/eurheartj/ehi818.
    1. van der Spoel T.I., Lee J.C., Vrijsen K., Sluijter J.P., Cramer M.J., Doevendans P.A., van Belle E., Chamuleau S.A. Non-surgical stem cell delivery strategies and in vivo cell tracking to injured myocardium. Int. J. Cardiovasc. Imaging. 2011;27:367–383. doi: 10.1007/s10554-010-9658-4.
    1. Roura S., Bago J.R., Soler-Botija C., Pujal J.M., Galvez-Monton C., Prat-Vidal C., Llucia-Valldeperas A., Blanco J., Bayes-Genis A. Human umbilical cord blood-derived mesenchymal stem cells promote vascular growth in vivo. PLoS One. 2012;7:e49447. doi: 10.1371/journal.pone.0049447.
    1. Kang B.J., Kim H., Lee S.K., Kim J., Shen Y., Jung S., Kang K.S., Im S.G., Lee S.Y., Choi M., et al. Umbilical-cord-blood-derived mesenchymal stem cells seeded onto fibronectin-immobilized polycaprolactone nanofiber improve cardiac function. Acta Biomater. 2014;10:3007–3017. doi: 10.1016/j.actbio.2014.03.013.
    1. Ries C., Egea V., Karow M., Kolb H., Jochum M., Neth P. MMP-2, MT1-MMP, and TIMP-2 are essential for the invasive capacity of human mesenchymal stem cells: differential regulation by inflammatory cytokines. Blood. 2007;109:4055–4063. doi: 10.1182/blood-2006-10-051060.
    1. De Becker A., Van Hummelen P., Bakkus M., Vande Broek I., De Wever J., De Waele M., Van Riet I. Migration of culture-expanded human mesenchymal stem cells through bone marrow endothelium is regulated by matrix metalloproteinase-2 and tissue inhibitor of metalloproteinase-3. Haematologica. 2007;92:440–449. doi: 10.3324/haematol.10475.
    1. Nomura T., Honmou O., Harada K., Houkin K., Hamada H., Kocsis J.D. I.V. infusion of brain-derived neurotrophic factor gene-modified human mesenchymal stem cells protects against injury in a cerebral ischemia model in adult rat. Neuroscience. 2005;136:161–169. doi: 10.1016/j.neuroscience.2005.06.062.
    1. Schrepfer S., Deuse T., Reichenspurner H., Fischbein M.P., Robbins R.C., Pelletier M.P. Stem cell transplantation: the lung barrier. Transplant Proc. 2007;39:573–576. doi: 10.1016/j.transproceed.2006.12.019.
    1. Ryu C.H., Park S.A., Kim S.M., Lim J.Y., Jeong C.H., Jun J.A., Oh J.H., Park S.H., Oh W.I., Jeun S.S. Migration of human umbilical cord blood mesenchymal stem cells mediated by stromal cell-derived factor-1/CXCR4 axis via Akt, ERK, and p38 signal transduction pathways. Biochem. Biophys. Res. Commun. 2010;398:105–110. doi: 10.1016/j.bbrc.2010.06.043.
    1. Thirumala S., Gimble J.M., Devireddy R.V. Cryopreservation of stromal vascular fraction of adipose tissue in a serum-free freezing medium. J. Tissue Eng. Regen. Med. 2010;4:224–232. doi: 10.1002/term.232.
    1. Thirumala S., Wu X., Gimble J.M., Devireddy R.V. Evaluation of polyvinylpyrrolidone as a cryoprotectant for adipose tissue-derived adult stem cells. Tissue Eng. Part C. 2010;16:783–92. doi: 10.1089/ten.tec.2009.0552.
    1. Fuller B.J. Cryoprotectants: the essential antifreezes to protect life in the frozen state. Cryo. Lett. 2004;25:375–388.
    1. Windrum P., Morris T.C., Drake M.B., Niederwieser D., Ruutu T., Subcommittee E.C.L.W.P.C. Variation in dimethyl sulfoxide use in stem cell transplantation: a survey of EBMT centres. Bone Marrow Transplant. 2005;36:601–603. doi: 10.1038/sj.bmt.1705100.
    1. Rowley S.D., Feng Z., Yadock D., Holmberg L., Macleod B., Heimfeld S. Post-thaw removal of DMSO does not completely abrogate infusional toxicity or the need for pre-infusion histamine blockade. Cytotherapy. 1999;1:439–446. doi: 10.1080/0032472031000141303.
    1. Zenhausern R., Tobler A., Leoncini L., Hess O.M., Ferrari P. Fatal cardiac arrhythmia after infusion of dimethyl sulfoxide-cryopreserved hematopoietic stem cells in a patient with severe primary cardiac amyloidosis and end-stage renal failure. Ann. Hematol. 2000;79:523–526. doi: 10.1007/s002770000186.
    1. Rodriguez L., Velasco B., Garcia J., Martin-Henao G.A. Evaluation of an automated cell processing device to reduce the dimethyl sulfoxide from hematopoietic grafts after thawing. Transfusion. 2005;45:1391–1397. doi: 10.1111/j.1537-2995.2005.00213.x.
    1. Park B.W., Jang S.J., Byun J.H., Kang Y.H., Choi M.J., Park W.U., Lee W.J., Rho G.J. Cryopreservation of human dental follicle tissue for use as a resource of autologous mesenchymal stem cells. J. Tissue Eng. Regen Med. 2014 doi: 10.1002/term.1945.
    1. Thirumala S., Zvonic S., Floyd E., Gimble J.M., Devireddy R.V. Effect of various freezing parameters on the immediate post-thaw membrane integrity of adipose tissue derived adult stem cells. Biotechnol. Prog. 2005;21:1511–1524. doi: 10.1021/bp050007q.
    1. Shu Z., Kang X., Chen H., Zhou X., Purtteman J., Yadock D., Heimfeld S., Gao D. Development of a reliable low-cost controlled cooling rate instrument for the cryopreservation of hematopoietic stem cells. Cytotherapy. 2010;12:161–169. doi: 10.3109/14653240903377037.
    1. Thirumala S., Goebel W.S., Woods E.J. Manufacturing and banking of mesenchymal stem cells. Expert Opin. Biol. Ther. 2013;13:673–691. doi: 10.1517/14712598.2013.763925.
    1. Baker K.F., Issacs J.D. Prospects for therapeutic tolerance in humans. Curr. Opin. Rheumatol. 2014;26:219–227. doi: 10.1097/BOR.0000000000000029.
    1. Wang Y., Wang F., Zhao H., Zhang X., Chen H., Zhang K. Human adipose-derived mesenchymal stem cells are resistant to HBV infection during differentiation into hepatocytes in vitro. Int. J. Mol. Sci. 2014;15:6096–6110. doi: 10.3390/ijms15046096.
    1. Choi Y.S., Dusting G.J., Stubbs S., Arunothayaraj S., Han X.L., Collas P., Morrison W.A., Dilley R.J. Differentiation of human adipose-derived stem cells into beating cardiomyocytes. J. Cell Mol. Med. 2010;14:878–889. doi: 10.1111/j.1582-4934.2010.01009.x.
    1. Timper K., Seboek D., Eberhardt M., Linscheid P., Christ-Crain M., Keller U., Muller B., Zulewski H. Human adipose tissue-derived mesenchymal stem cells differentiate into insulin, somatostatin, and glucagon expressing cells. Biochem. Biophys. Res. Commun. 2006;341:1135–1140. doi: 10.1016/j.bbrc.2006.01.072.
    1. Anzalone R., Lo Iacono M., Corrao S., Magno F., Loria T., Cappello F., Zummo G., Farina F., La Rocca G. New emerging potentials for human Wharton's jelly mesenchymal stem cells: immunological features and hepatocyte-like differentiative capacity. Stem Cells Dev. 2010;19:423–438. doi: 10.1089/scd.2009.0299.
    1. Mitchell K.E., Weiss M.L., Mitchell B.M., Martin P., Davis D., Morales L., Helwig B., Beerenstrauch M., Abou-Easa K., Hildreth T., et al. Matrix cells from Wharton's jelly form neurons and glia. Stem Cells. 2003;21:50–60. doi: 10.1634/stemcells.21-1-50.

Source: PubMed

3
Abonner