Phenotype, donor age and gender affect function of human bone marrow-derived mesenchymal stromal cells

Georg Siegel, Torsten Kluba, Ursula Hermanutz-Klein, Karen Bieback, Hinnak Northoff, Richard Schäfer, Georg Siegel, Torsten Kluba, Ursula Hermanutz-Klein, Karen Bieback, Hinnak Northoff, Richard Schäfer

Abstract

Background: Mesenchymal stromal cells (MSCs) are attractive for cell-based therapies ranging from regenerative medicine and tissue engineering to immunomodulation. However, clinical efficacy is variable and it is unclear how the phenotypes defining bone marrow (BM)-derived MSCs as well as donor characteristics affect their functional properties.

Methods: BM-MSCs were isolated from 53 (25 female, 28 male; age: 13 to 80 years) donors and analyzed by: (1) phenotype using flow cytometry and cell size measurement; (2) in vitro growth kinetics using population doubling time; (3) colony formation capacity and telomerase activity; and (4) function by in vitro differentiation capacity, suppression of T cell proliferation, cytokines and trophic factors secretion, and hormone and growth factor receptor expression. Additionally, expression of Oct4, Nanog, Prdm14 and SOX2 mRNA was compared to pluripotent stem cells.

Results: BM-MSCs from younger donors showed increased expression of MCAM, VCAM-1, ALCAM, PDGFRβ, PDL-1, Thy1 and CD71, and led to lower IL-6 production when co-cultured with activated T cells. Female BM-MSCs showed increased expression of IFN-γR1 and IL-6β, and were more potent in T cell proliferation suppression. High-clonogenic BM-MSCs were smaller, divided more rapidly and were more frequent in BM-MSC preparations from younger female donors. CD10, β1integrin, HCAM, CD71, VCAM-1, IFN-γR1, MCAM, ALCAM, LNGFR and HLA ABC were correlated to BM-MSC preparations with high clonogenic potential and expression of IFN-γR1, MCAM and HLA ABC was associated with rapid growth of BM-MSCs. The mesodermal differentiation capacity of BM-MSCs was unaffected by donor age or gender but was affected by phenotype (CD10, IFN-γR1, GD2). BM-MSCs from female and male donors expressed androgen receptor and FGFR3, and secreted VEGF-A, HGF, LIF, Angiopoietin-1, basic fibroblast growth factor (bFGF) and NGFB. HGF secretion correlated negatively to the expression of CD71, CD140b and Galectin 1. The expression of Oct4, Nanog and Prdm14 mRNA in BM-MSCs was much lower compared to pluripotent stem cells and was not related to donor age or gender. Prdm14 mRNA expression correlated positively to the clonogenic potential of BM-MSCs.

Conclusions: By identifying donor-related effects and assigning phenotypes of BM-MSC preparations to functional properties, we provide useful tools for assay development and production for clinical applications of BM-MSC preparations.

Figures

Figure 1
Figure 1
Flow cytometric analyses. The expression of CD10, CD14, CD19, CD29, CD31, CD34, CD43, CD44, CD45, CD56, CD59, CD71, CD73, CD80, CD86, CD90, CD93, CD105, CD106, CD119, CD130, CD133, CD140a, CD140b, CD146, CD173, CD166, CD243, CD271, CD273, CD274, Galectin 1, GD2, MSCA-1, SSEA-1, SSEA-4 and HLA class I was analyzed by flow cytometry in MSC preparations from multiple donors identifying three groups of marker expression. Markers that were expressed on all/most of the cells or on none/very few of the cells within the respective MSC preparation (A) and markers that identified MSC subpopulations by presence or absence of the respective marker (B) (n = 33 except for CD80 (n = 30), CD86 (n = 30), CD119 (n = 27), CD130 (n = 34), CD273 (n = 30), CD274 (n = 30), Galectin 1 (n = 24), and SSEA-4 (n = 36)). Analyses of specific antibody mediated fluorescence per cell (ΔGeo Mean) identified markers that were expressed at high and low density per cell (C, D) (n = 36 except for CD80 (n = 32), CD86 (n = 32), CD119 (n = 29), CD130 (n = 37), CD273 (n = 32), CD274 (n = 32), Galectin 1 (n = 26), and SSEA-4 (n = 39)). ANOVA analysis of variance followed by Tukey’s Multiple Comparison Test (***P <0.001). Error bars: SD.
Figure 2
Figure 2
Correlation analyses of marker expression to donor age and gender distribution. The percentage of CD71+, CD146+, and CD274+ cells and the specific antibody mediated fluorescence per cell (ΔGeo Mean) of CD71, CD90, CD106, CD140b, CD146, CD166 and CD274 correlated negatively with the donor age (n = 34 except for CD274 (n = 31)) (A-J). Spearman two-tailed correlation test (*P <0.05; **P <0.01). MSC preparations from female donors harbored significantly more CD119+ cells (n = 11) and CD130+ cells (n = 16) compared to MSC preparations from male donors (CD119+ cells: n = 17; CD130+ cells: n = 19) (K, L). Two-tailed Student’s t-test (*P <0.05; **P <0.01). Error bars: SD.
Figure 3
Figure 3
Analyses of cell size and growth kinetics. The diameter of MSCs from female donors (n = 14) was slightly but significantly smaller compared to male donors (n = 11) (A). The PDT of MSCs from female donors (n = 25) was significantly reduced compared to MSCs from male donors (n = 27) (B). Two-tailed Student’s t-test (*P <0.05). Error bars: SD. The cell diameter correlated positively to the PDT (n = 25) (C). No correlation was found between the donor age and the proliferation capacity of the MSCs (n = 52) (D) and between the donor age to the cell size (n = 25) (E). The specific antibody mediated fluorescence per cell (ΔGeo Mean) of CD105, CD119, CD140a, CD166 and HLA ABC correlated negatively with the cell size (n = 22 except for CD119 (n = 17)) (F-J). The specific antibody mediated fluorescence per cell (ΔGeo Mean) of CD105, CD119, and HLA ABC correlated negatively with the PDT (n = 27 except for CD119 (n = 21)) (K-M). The PDT correlated negatively with the percentage of CD146+ cells within the MSC preparations (n = 27) (N). Spearman two-tailed correlation test (*P <0.05; **P <0.01; ***P <0.001; ****P <0.0001).
Figure 4
Figure 4
Analyses of clonogenic potential. Comparing all ages and both genders, MSC preparations from the BM of younger donors trended toward more colony forming cells than MSC preparations from older donors (n = 50) (A). Spearman two-tailed correlation test. Comparing age groups including both genders, significantly more colonies could be detected in young donors (<45 years) (n = 11) compared to middle-aged (n = 26) and older donors (n = 13) (B). ANOVA analysis of variance followed by Tukey’s Multiple Comparison Test (*P <0.05; **P <0.01). Error bars: SD. In BM-MSC preparations from female donors more colony forming cells could be detected (n = 23) compared to BM-MSC preparations from male donors (n = 27) (C). Two-tailed Student’s t-test (*P <0.05). Error bars: SD. Age-group specific analyses for each gender confirmed that more colonies could be detected in young female donors (<45 years) compared to older donors (D, E). ANOVA analysis of variance followed by Tukey’s Multiple Comparison Test (*P <0.05). Error bars: SD. The clonogenic potential of the cells within the BM-MSC preparations was negatively correlated to the population doubling time (n = 42) and the cell diameter (n = 22) (F, G). Spearman two-tailed correlation test (**P <0.01; ****P <0.0001). The percentage of CD10+, CD71+, CD106+, CD119+ CD146+, CD166+ and CD271+ cells correlated positively with the clonogenic potential of the BM-MSCs (n = 31 except for CD119 (n = 25)) (H-N). The specific antibody mediated fluorescence per cell (ΔGeo Mean) of CD29, CD44, CD119, CD146, CD166 and HLA ABC correlated positively with the clonogenic potential of the MSCs (n = 31 except for CD119 (n = 25)) (O-T). Spearman two-tailed correlation test (*P <0.05; **P <0.01; ***P <0.001; ****P <0.0001).
Figure 5
Figure 5
Analyses of the immunosuppressive potential of MSCs. MSCs from both female and male donors significantly suppressed the proliferation of activated allogeneic T cells. MSCs from female donors (n = 18) showed a significantly higher potency to suppress T cell proliferation than MSCs from male donors (n = 16). Data are normalized to the proliferation index of PBMNCs alone (A). ANOVA analysis of variance followed by Tukey’s Multiple Comparison Test (*P <0.05; ***P <0.001). Error bars: SD. No correlation of the donor age to the potential to suppress T cell proliferation could be detected (B). Upon co-culturing of MSCs from older donors with activated allogeneic T cells more IL-6 could be detected in the supernatant compared to MSCs from younger donors that were co-cultured with activated T cells (n = 17) (C). Spearman two-tailed correlation test (*P <0.05). Although a trend toward a negative correlation was observed, no significant correlation of IDO 1 mRNA expression to T cell proliferation could be detected (n = 8) (D). Spearman two-tailed correlation test. No difference of IDO 1 mRNA expression from male MSC donors compared to female MSC donors could be detected (n = 22) (E). Two-tailed Student’s t-test. Error bars: SD. No correlation of the IDO 1 mRNA expression to the donor age could be observed (n = 22) (F). Spearman two-tailed correlation test.
Figure 6
Figure 6
Mesodermal differentiation potential of MSCs. No donor age or gender related differences could be detected for the adipogenic, osteogenic and chondrogenic in vitro differentiation capacity of the MSC preparations as analysed by lineage specific staining (Oil Red O for adipogensis; Alizarin Red for osteogenesis, and Safranain O for chondrogenesis (n = 40); differentiation: A, C, E; negative controls: B, D) and no statistically significant differences could be detected for the lineage specific mRNA expression (LPL (n = 44) and PPARγ (n = 48) for adipogenesis; OPN (n = 17) and AP (n = 41) for osteogenesis; SOX9 (n = 47) and COLL2 (n = 32) for chondrogenesis; F-Q). Two-tailed Student’s t-test and Spearman two-tailed correlation test. Error bars: SD. Scale bars: 100 μm.
Figure 7
Figure 7
Expression of Oct4, Nanog and Prdm14 mRNA and functional relevance. The MSCs expressed significantly more Oct4 mRNA than Nanog or Prdm14 mRNA; however, no significant difference in Nanog mRNA expression compared to Prdm14 mRNA expression could be detected (n = 28 except for Prdm14 (n = 23)) (A). Compared to pluripotent HUES9 cells and pluripotent NCCIT cells the expression of Oct4 mRNA, Nanog mRNA and Prdm14 mRNA was much lower in MSCs (B-D). Compared to NCCIT cells the HUES9 cells expressed more Oct4 mRNA, Nanog mRNA and Prdm14 mRNA (B-D). ANOVA analysis of variance followed by Tukey’s Multiple Comparison Test (*P <0.05; ***P <0.001). Error bars: SD. No correlation of the expression of Oct4, Nanog and Prdm14 mRNA to donor age (n = 28 except for Prdm14 (n = 23)) (E-G) or any gender-related differences could be detected (n = 28 except for Prdm14 (n = 23)) (H-J). The clonogenic potential of MSCs correlated positively to the expression of Prdm14 mRNA (n = 18) but not to Oct4 (n = 24) or Nanog mRNA (n = 24) (K-M). Two-tailed Student’s t-test and Spearman two-tailed correlation test (**P <0.01). Error bars: SD.

References

    1. Friedenstein AJ, Petrakova KV, Kurolesova AI, Frolova GP. Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation. 1968;6:230–247. doi: 10.1097/00007890-196803000-00009.
    1. Friedenstein AJ, Chailakhyan RK, Latsinik NV, Panasyuk AF, Keiliss-Borok IV. Stromal cells responsible for transferring the microenvironment of the hemopoietic tissues. Cloning in vitro and retransplantation in vivo. Transplantation. 1974;17:331–340. doi: 10.1097/00007890-197404000-00001.
    1. Muguruma Y, Yahata T, Miyatake H, Sato T, Uno T, Itoh J, Kato S, Ito M, Hotta T, Ando K. Reconstitution of the functional human hematopoietic microenvironment derived from human mesenchymal stem cells in the murine bone marrow compartment. Blood. 2006;107:1878–1887. doi: 10.1182/blood-2005-06-2211.
    1. Miura Y, Miura M, Gronthos S, Allen MR, Cao C, Uveges TE, Bi Y, Ehirchiou D, Kortesidis A, Shi S, Zhang L. Defective osteogenesis of the stromal stem cells predisposes CD18-null mice to osteoporosis. Proc Natl Acad Sci U S A. 2005;102:14022–14027. doi: 10.1073/pnas.0409397102.
    1. Horwitz EM, Gordon PL, Koo WK, Marx JC, Neel MD, McNall RY, Muul L, Hofmann T. Isolated allogeneic bone marrow-derived mesenchymal cells engraft and stimulate growth in children with osteogenesis imperfecta: Implications for cell therapy of bone. Proc Natl Acad Sci U S A. 2002;99:8932–8937. doi: 10.1073/pnas.132252399.
    1. Le Blanc K, Frassoni F, Ball L, Locatelli F, Roelofs H, Lewis I, Lanino E, Sundberg B, Bernardo ME, Remberger M, Dini G, Egeler RM, Bacigalupo A, Fibbe W, Ringdén O. Developmental Committee of the European Group for Blood and Marrow Transplantation. Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet. 2008;371:1579–1586. doi: 10.1016/S0140-6736(08)60690-X.
    1. Williams AR, Hare JM. Mesenchymal stem cells: biology, pathophysiology, translational findings, and therapeutic implications for cardiac disease. Circ Res. 2011;109:923–940. doi: 10.1161/CIRCRESAHA.111.243147.
    1. Duijvestein M, Vos AC, Roelofs H, Wildenberg ME, Wendrich BB, Verspaget HW, Kooy-Winkelaar EM, Koning F, Zwaginga JJ, Fidder HH, Verhaar AP, Fibbe WE, van den Brink GR, Hommes DW. Autologous bone marrow-derived mesenchymal stromal cell treatment for refractory luminal Crohn’s disease: results of a phase I study. Gut. 2010;59:1662–1669. doi: 10.1136/gut.2010.215152.
    1. Freedman MS, Bar-Or A, Atkins HL, Karussis D, Frassoni F, Lazarus H, Scolding N, Slavin S, Le Blanc K, Uccelli A. The therapeutic potential of mesenchymal stem cell transplantation as a treatment for multiple sclerosis: consensus report of the international MSCT study group. Mult Scler. 2010;16:503–510. doi: 10.1177/1352458509359727.
    1. Phinney DG, Prockop DJ. Concise review: mesenchymal stem/multipotent stromal cells: the state of transdifferentiation and modes of tissue repair–current views. Stem Cells. 2007;25:2896–2902. doi: 10.1634/stemcells.2007-0637.
    1. Phinney DG, Kopen G, Righter W, Webster S, Tremain N, Prockop DJ. Donor variation in the growth properties and osteogenic potential of human marrow stromal cells. J Cell Biochem. 1999;75:424–436. doi: 10.1002/(SICI)1097-4644(19991201)75:3<424::AID-JCB8>;2-8.
    1. Russell KC, Phinney DG, Lacey MR, Barrilleaux BL, Meyertholen KE, O’Connor KC. In vitro high-capacity assay to quantify the clonal heterogeneity in trilineage potential of mesenchymal stem cells reveals a complex hierarchy of lineage commitment. Stem Cells. 2010;28:788–798. doi: 10.1002/stem.312.
    1. Quirici N, Soligo D, Bossolasco P, Servida F, Lumini C, Deliliers GL. Isolation of bone marrow mesenchymal stem cells by anti-nerve growth factor receptor antibodies. Exp Hematol. 2002;30:783–791. doi: 10.1016/S0301-472X(02)00812-3.
    1. Schäfer R, Schnaidt M, Klaffschenkel RA, Siegel G, Schüle M, Rädlein MA, Hermanutz-Klein U, Ayturan M, Buadze M, Gassner C, Danielyan L, Kluba T, Northoff H, Flegel WA. Expression of blood group genes by mesenchymal stem cells. Br J Haematol. 2011;153:520–528. doi: 10.1111/j.1365-2141.2011.08652.x.
    1. Schäfer R, Dominici M, Müller I, Horwitz E, Asahara T, Bulte JW, Bieback K, Le Blanc K, Bühring HJ, Capogrossi MC, Dazzi F, Gorodetsky R, Henschler R, Handgretinger R, Kajstura J, Kluger PJ, Lange C, Luettichau I, Mertsching H, Schrezenmeier H, Sievert KD, Strunk D, Verfaillie C, Northoff H. Basic research and clinical applications of non-hematopoietic stem cells, 4–5 April 2008, Tubingen, Germany. Cytotherapy. 2009;11:245–255. doi: 10.1080/14653240802582117.
    1. Bianco P, Barker R, Brustle O, Cattaneo E, Clevers H, Daley GQ, De Luca M, Goldstein L, Lindvall O, Mummery C, Robey PG, Sattler de Sousa E Brito C, Smith A. Regulation of stem cell therapies under attack in Europe: for whom the bell tolls. EMBO J. 2013. Epub ahead of print.
    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. Siegel G, Schäfer R, Dazzi F. The immunosuppressive properties of mesenchymal stem cells. Transplantation. 2009;87(9 Suppl):S45–S49.
    1. Siegel G, Krause P, Wohrle S, Nowak P, Ayturan M, Kluba T, Brehm BR, Neumeister B, Kohler D, Rosenberger P, Just L, Northoff H, Schäfer R. Bone marrow-derived human mesenchymal stem cells express cardiomyogenic proteins but do not exhibit functional cardiomyogenic differentiation potential. Stem Cells Dev. 2012;21:2457–2470. doi: 10.1089/scd.2011.0626.
    1. Song L, Tuan RS. Transdifferentiation potential of human mesenchymal stem cells derived from bone marrow. FASEB J. 2004;18:980–982.
    1. Caplan AI, Dennis JE. Mesenchymal stem cells as trophic mediators. J Cell Biochem. 2006;98:1076–1084. doi: 10.1002/jcb.20886.
    1. Doorn J, Moll G, Le Blanc K, van Blitterswijk C, de Boer J. Therapeutic applications of mesenchymal stromal cells: paracrine effects and potential improvements. Tissue Eng Part B Rev. 2012;18:101–115. doi: 10.1089/ten.teb.2011.0488.
    1. Cox G, Boxall SA, Giannoudis PV, Buckley CT, Roshdy T, Churchman SM, McGonagle D, Jones E. High abundance of CD271(+) multipotential stromal cells (MSCs) in intramedullary cavities of long bones. Bone. 2012;50:510–517. doi: 10.1016/j.bone.2011.07.016.
    1. Churchman SM, Ponchel F, Boxall SA, Cuthbert R, Kouroupis D, Roshdy T, Giannoudis PV, Emery P, McGonagle D, Jones EA. Transcriptional profile of native CD271+ multipotential stromal cells: evidence for multiple fates, with prominent osteogenic and Wnt pathway signaling activity. Arthritis Rheum. 2012;64:2632–2643. doi: 10.1002/art.34434.
    1. Dominici M, Paolucci P, Conte P, Horwitz EM. Heterogeneity of multipotent mesenchymal stromal cells: from stromal cells to stem cells and vice versa. Transplantation. 2009;87(9 Suppl):S36–S42.
    1. Muraglia A, Cancedda R, Quarto R. Clonal mesenchymal progenitors from human bone marrow differentiate in vitro according to a hierarchical model. J Cell Sci. 2000;113:1161–1166.
    1. Russell KC, Lacey MR, Gilliam JK, Tucker HA, Phinney DG, O’Connor KC. Clonal analysis of the proliferation potential of human bone marrow mesenchymal stem cells as a function of potency. Biotechnol Bioeng. 2011;108:2716–2726. doi: 10.1002/bit.23193.
    1. da Silva ML, Caplan AI, Nardi NB. In search of the in vivo identity of mesenchymal stem cells. Stem Cells. 2008;26:2287–2299. doi: 10.1634/stemcells.2007-1122.
    1. Jones E, McGonagle D. Human bone marrow mesenchymal stem cells in vivo. Rheumatology (Oxford) 2008;47:126–131.
    1. Battula VL, Treml S, Bareiss PM, Gieseke F, Roelofs H, de Zwart P, Muller I, Schewe B, Skutella T, Fibbe WE, Kanz L, Bühring HJ. Isolation of functionally distinct mesenchymal stem cell subsets using antibodies against CD56, CD271, and mesenchymal stem cell antigen-1. Haematologica. 2009;94:173–184. doi: 10.3324/haematol.13740.
    1. Bühring HJ, Battula VL, Treml S, Schewe B, Kanz L, Vogel W. Novel markers for the prospective isolation of human MSC. Ann N Y Acad Sci. 2007;1106:262–271. doi: 10.1196/annals.1392.000.
    1. Martinez C, Hofmann TJ, Marino R, Dominici M, Horwitz EM. Human bone marrow mesenchymal stromal cells express the neural ganglioside GD2: a novel surface marker for the identification of MSCs. Blood. 2007;109:4245–4248. doi: 10.1182/blood-2006-08-039347.
    1. Schäfer R. Does the adult stroma contain stem cells? Adv Biochem Eng Biotechnol. 2013;129:177–189.
    1. Phinney DG. Functional heterogeneity of mesenchymal stem cells: implications for cell therapy. J Cell Biochem. 2012;113:2806–2812. doi: 10.1002/jcb.24166.
    1. Buick RN, MacKillop WJ. Measurement of self-renewal in culture of clonogenic cells from human ovarian carcinoma. Br J Cancer. 1981;44:349–355. doi: 10.1038/bjc.1981.191.
    1. Thomson SP, Meyskens FL Jr. Method for measurement of self-renewal capacity of clonogenic cells from biopsies of metastatic human malignant melanoma. Cancer Res. 1982;42:4606–4613.
    1. Rose RA, Jiang H, Wang X, Helke S, Tsoporis JN, Gong N, Keating SC, Parker TG, Backx PH, Keating A. Bone marrow-derived mesenchymal stromal cells express cardiac-specific markers, retain the stromal phenotype, and do not become functional cardiomyocytes in vitro. Stem Cells. 2008;26:2884–2892. doi: 10.1634/stemcells.2008-0329.
    1. Henderson JK, Draper JS, Baillie HS, Fishel S, Thomson JA, Moore H, Andrews PW. Preimplantation human embryos and embryonic stem cells show comparable expression of stage-specific embryonic antigens. Stem Cells. 2002;20:329–337. doi: 10.1634/stemcells.20-4-329.
    1. Tsuneyoshi N, Sumi T, Onda H, Nojima H, Nakatsuji N, Suemori H. PRDM14 suppresses expression of differentiation marker genes in human embryonic stem cells. Biochem Biophys Res Commun. 2008;367:899–905. doi: 10.1016/j.bbrc.2007.12.189.
    1. Battula VL, Bareiss PM, Treml S, Conrad S, Albert I, Hojak S, Abele H, Schewe B, Just L, Skutella T, Bühring HJ. Human placenta and bone marrow derived MSC cultured in serum-free, b-FGF-containing medium express cell surface frizzled-9 and SSEA-4 and give rise to multilineage differentiation. Differentiation. 2007;75:279–291. doi: 10.1111/j.1432-0436.2006.00139.x.
    1. Fazzi R, Pacini S, Carnicelli V, Trombi L, Montali M, Lazzarini E, Petrini M. Mesodermal progenitor cells (MPCs) differentiate into mesenchymal stromal cells (MSCs) by activation of Wnt5/calmodulin signalling pathway. PLoS One. 2011;6:e25600. doi: 10.1371/journal.pone.0025600.
    1. Roobrouck VD, Clavel C, Jacobs SA, Ulloa-Montoya F, Crippa S, Sohni A, Roberts SJ, Luyten FP, Van Gool SW, Sampaolesi M, Delforge M, Luttun A, Verfaillie CM. Differentiation potential of human postnatal mesenchymal stem cells, mesoangioblasts, and multipotent adult progenitor cells reflected in their transcriptome and partially influenced by the culture conditions. Stem Cells. 2011;29:871–882. doi: 10.1002/stem.633.
    1. Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz-Gonzalez XR, Reyes M, Lenvik T, Lund T, Blackstad M, Du J, Aldrich S, Lisberg A, Low WC, Largaespada DA, Verfaillie CM. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature. 2002;418:41–49. doi: 10.1038/nature00870. Erratum in: Nature 2007, 447:879–880.
    1. Serafini M, Dylla SJ, Oki M, Heremans Y, Tolar J, Jiang Y, Buckley SM, Pelacho B, Burns TC, Frommer S, Rossi DJ, Bryder D, Panoskaltsis-Mortari A, O'Shaughnessy MJ, Nelson-Holte M, Fine GC, Weissman IL, Blazar BR, Verfaillie CM. Hematopoietic reconstitution by multipotent adult progenitor cells: precursors to long-term hematopoietic stem cells. J Exp Med. 2007;204:129–139. doi: 10.1084/jem.20061115.
    1. D’Ippolito G, Diabira S, Howard GA, Menei P, Roos BA, Schiller PC. Marrow-isolated adult multilineage inducible (MIAMI) cells, a unique population of postnatal young and old human cells with extensive expansion and differentiation potential. J Cell Sci. 2004;117:2971–2981. doi: 10.1242/jcs.01103.
    1. Kucia M, Halasa M, Wysoczynski M, Baskiewicz-Masiuk M, Moldenhawer S, Zuba-Surma E, Czajka R, Wojakowski W, Machalinski B, Ratajczak MZ. Morphological and molecular characterization of novel population of CXCR4+ SSEA-4+ Oct-4+ very small embryonic-like cells purified from human cord blood: preliminary report. Leukemia. 2007;21:297–303. doi: 10.1038/sj.leu.2404470.
    1. Kucia M, Reca R, Campbell FR, Zuba-Surma E, Majka M, Ratajczak J, Ratajczak MZ. A population of very small embryonic-like (VSEL) CXCR4(+)SSEA-1(+)Oct-4+ stem cells identified in adult bone marrow. Leukemia. 2006;20:857–869. doi: 10.1038/sj.leu.2404171.
    1. Rasini V, Dominici M, Kluba T, Siegel G, Lusenti G, Northoff H, Horwitz EM, Schäfer R. Mesenchymal stromal/stem cells markers in the human bone marrow. Cytotherapy. 2013;15:292–306. doi: 10.1016/j.jcyt.2012.11.009.
    1. Ren G, Zhang L, Zhao X, Xu G, Zhang Y, Roberts AI, Zhao RC, Shi Y. Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell. 2008;2:141–150. doi: 10.1016/j.stem.2007.11.014.
    1. Le Blanc K, Rasmusson I, Sundberg B, Gotherstrom C, Hassan M, Uzunel M, Ringden O. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet. 2004;363:1439–1441. doi: 10.1016/S0140-6736(04)16104-7.
    1. Connick P, Kolappan M, Crawley C, Webber DJ, Patani R, Michell AW, Du MQ, Luan SL, Altmann DR, Thompson AJ, Compston A, Scott MA, Miller DH, Chandran S. Autologous mesenchymal stem cells for the treatment of secondary progressive multiple sclerosis: an open-label phase 2a proof-of-concept study. Lancet Neurol. 2012;11:150–156. doi: 10.1016/S1474-4422(11)70305-2.
    1. Connick P, Kolappan M, Patani R, Scott MA, Crawley C, He XL, Richardson K, Barber K, Webber DJ, Wheeler-Kingshott CA, Tozer DJ, Samson RS, Thomas DL, Du MQ, Luan SL, Michell AW, Altmann DR, Thompson AJ, Miller DH, Compston A, Chandran S. The mesenchymal stem cells in multiple sclerosis (MSCIMS) trial protocol and baseline cohort characteristics: an open-label pre-test: post-test study with blinded outcome assessments. Trials. 2011;12:62. doi: 10.1186/1745-6215-12-62.
    1. Löb S, Konigsrainer A, Schäfer R, Rammensee HG, Opelz G, Terness P. Levo- but not dextro-1-methyl tryptophan abrogates the IDO activity of human dendritic cells. Blood. 2008;111:2152–2154. doi: 10.1182/blood-2007-10-116111.
    1. Francois M, Romieu-Mourez R, Li M, Galipeau J. Human MSC suppression correlates with cytokine induction of indoleamine 2,3-dioxygenase and bystander M2 macrophage differentiation. Mol Ther. 2012;20:187–195. doi: 10.1038/mt.2011.189.
    1. Gieseke F, Bohringer J, Bussolari R, Dominici M, Handgretinger R, Müller I. Human multipotent mesenchymal stromal cells use galectin-1 to inhibit immune effector cells. Blood. 2010;116:3770–3779. doi: 10.1182/blood-2010-02-270777.
    1. Mourcin F, Breton C, Tellier J, Narang P, Chasson L, Jorquera A, Coles M, Schiff C, Mancini SJ. Galectin-1-expressing stromal cells constitute a specific niche for pre-BII cell development in mouse bone marrow. Blood. 2011;117:6552–6561. doi: 10.1182/blood-2010-12-323113.
    1. Fife BT, Pauken KE. The role of the PD-1 pathway in autoimmunity and peripheral tolerance. Ann N Y Acad Sci. 2011;1217:45–59. doi: 10.1111/j.1749-6632.2010.05919.x.
    1. Augello A, Tasso R, Negrini SM, Amateis A, Indiveri F, Cancedda R, Pennesi G. Bone marrow mesenchymal progenitor cells inhibit lymphocyte proliferation by activation of the programmed death 1 pathway. Eur J Immunol. 2005;35:1482–1490. doi: 10.1002/eji.200425405.
    1. Fiorina P, Jurewicz M, Augello A, Vergani A, Dada S, La Rosa S, Selig M, Godwin J, Law K, Placidi C, Smith RN, Capella C, Rodig S, Adra CN, Atkinson M, Sayegh MH, Abdi R. Immunomodulatory function of bone marrow-derived mesenchymal stem cells in experimental autoimmune type 1 diabetes. J Immunol. 2009;183:993–1004. doi: 10.4049/jimmunol.0900803.
    1. Choumerianou DM, Martimianaki G, Stiakaki E, Kalmanti L, Kalmanti M, Dimitriou H. Comparative study of stemness characteristics of mesenchymal cells from bone marrow of children and adults. Cytotherapy. 2010;12:881–887. doi: 10.3109/14653249.2010.501790.
    1. Alves H, van Ginkel J, Groen N, Hulsman M, Mentink A, Reinders M, van Blitterswijk C, de Boer J. A mesenchymal stromal cell gene signature for donor age. PLoS One. 2012;7:e42908. doi: 10.1371/journal.pone.0042908.
    1. Kurozumi K, Nakamura K, Tamiya T, Kawano Y, Ishii K, Kobune M, Hirai S, Uchida H, Sasaki K, Ito Y, Kato K, Honmou O, Houkin K, Date I, Hamada H. Mesenchymal stem cells that produce neurotrophic factors reduce ischemic damage in the rat middle cerebral artery occlusion model. Mol Ther. 2005;11:96–104.
    1. Nomura T, Honmou O, Harada K, Houkin K, Hamada H, Kocsis JD. 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. Nguyen BK, Maltais S, Perrault LP, Tanguay JF, Tardif JC, Stevens LM, Borie M, Harel F, Mansour S, Noiseux N. Improved function and myocardial repair of infarcted heart by intracoronary injection of mesenchymal stem cell-derived growth factors. J Cardiovasc Transl Res. 2010;3:547–558. doi: 10.1007/s12265-010-9171-0.
    1. van Poll D, Parekkadan B, Cho CH, Berthiaume F, Nahmias Y, Tilles AW, Yarmush ML. Mesenchymal stem cell-derived molecules directly modulate hepatocellular death and regeneration in vitro and in vivo. Hepatology. 2008;47:1634–1643. doi: 10.1002/hep.22236.
    1. Lee JW, Fang X, Krasnodembskaya A, Howard JP, Matthay MA. 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. Crisostomo PR, Wang M, Herring CM, Morrell ED, Seshadri P, Meldrum KK, Meldrum DR. Sex dimorphisms in activated mesenchymal stem cell function. Shock. 2006;26:571–574. doi: 10.1097/01.shk.0000233195.63859.ef.
    1. Crisostomo PR, Wang M, Herring CM, Markel TA, Meldrum KK, Lillemoe KD, Meldrum DR. Gender differences in injury induced mesenchymal stem cell apoptosis and VEGF, TNF, IL-6 expression: role of the 55 kDa TNF receptor (TNFR1) J Mol Cell Cardiol. 2007;42:142–149. doi: 10.1016/j.yjmcc.2006.09.016.
    1. Chen DF, Du SH, Zhang HL, Li H, Zhou JH, Li YW, Yi XH, Hou QK, Wu J, Zeng HP, Hua ZC. Autocrine BMP4 signaling involves effect of cholesterol myristate on proliferation of mesenchymal stem cells. Steroids. 2009;74:1066–1072. doi: 10.1016/j.steroids.2009.08.008.
    1. Wislet-Gendebien S, Bruyere F, Hans G, Leprince P, Moonen G, Rogister B. Nestin-positive mesenchymal stem cells favour the astroglial lineage in neural progenitors and stem cells by releasing active BMP4. BMC Neurosci. 2004;5:33. doi: 10.1186/1471-2202-5-33.
    1. Ng F, Boucher S, Koh S, Sastry KS, Chase L, Lakshmipathy U, Choong C, Yang Z, Vemuri MC, Rao MS, Tanavde V. PDGF, TGF-beta, and FGF signaling is important for differentiation and growth of mesenchymal stem cells (MSCs): transcriptional profiling can identify markers and signaling pathways important in differentiation of MSCs into adipogenic, chondrogenic, and osteogenic lineages. Blood. 2008;112:295–307. doi: 10.1182/blood-2007-07-103697.
    1. Fekete N, Gadelorge M, Fürst D, Maurer C, Dausend J, Fleury-Cappellesso S, Mailänder V, Lotfi R, Ignatius A, Sensebé L, Bourin P, Schrezenmeier H, Rojewski MT. Platelet lysate from whole blood-derived pooled platelet concentrates and apheresis-derived platelet concentrates for the isolation and expansion of human bone marrow mesenchymal stromal cells: production process, content and identification of active components. Cytotherapy. 2012;14:540–554. doi: 10.3109/14653249.2012.655420.
    1. Kim JH, Lee MC, Seong SC, Park KH, Lee S. Enhanced proliferation and chondrogenic differentiation of human synovium-derived stem cells expanded with basic fibroblast growth factor. Tissue Eng Part A. 2011;17:991–1002. doi: 10.1089/ten.tea.2010.0277.
    1. Lai WT, Krishnappa V, Phinney DG. Fibroblast growth factor 2 (Fgf2) inhibits differentiation of mesenchymal stem cells by inducing Twist2 and Spry4, blocking extracellular regulated kinase activation, and altering Fgf receptor expression levels. Stem Cells. 2011;29:1102–1111. doi: 10.1002/stem.661.
    1. Coutu DL, Francois M, Galipeau J. Inhibition of cellular senescence by developmentally regulated FGF receptors in mesenchymal stem cells. Blood. 2011;117:6801–6812. doi: 10.1182/blood-2010-12-321539.
    1. Gupta V, Bhasin S, Guo W, Singh R, Miki R, Chauhan P, Choong K, Tchkonia T, Lebrasseur NK, Flanagan JN, Hamilton JA, Viereck JC, Narula NS, Kirkland JL, Jasuja R. Effects of dihydrotestosterone on differentiation and proliferation of human mesenchymal stem cells and preadipocytes. Mol Cell Endocrinol. 2008;296:32–40. doi: 10.1016/j.mce.2008.08.019.
    1. Breu A, Sprinzing B, Merkl K, Bechmann V, Kujat R, Jenei-Lanzl Z, Prantl L, Angele P. Estrogen reduces cellular aging in human mesenchymal stem cells and chondrocytes. J Orthop Res. 2011;29:1563–1571. doi: 10.1002/jor.21424.

Source: PubMed

3
Tilaa