In vivo efficacy of endothelial growth medium stimulated mesenchymal stem cells derived from patients with critical limb ischemia

Rida Al-Rifai, Philippe Nguyen, Nicole Bouland, Christine Terryn, Lukshe Kanagaratnam, Gaël Poitevin, Caroline François, Catherine Boisson-Vidal, Marie-Antoinette Sevestre, Claire Tournois, Rida Al-Rifai, Philippe Nguyen, Nicole Bouland, Christine Terryn, Lukshe Kanagaratnam, Gaël Poitevin, Caroline François, Catherine Boisson-Vidal, Marie-Antoinette Sevestre, Claire Tournois

Abstract

Background: Cell therapy has been proposed for patients with critical limb ischemia (CLI). Autologous bone marrow derived cells (BMCs) have been mostly used, mesenchymal stem cells (MSCs) being an alternative. The aim of this study was to characterize two types of MSCs and evaluate their efficacy.

Methods: MSCs were obtained from CLI-patients BMCs. Stimulated- (S-) MSCs were cultured in endothelial growth medium. Cells were characterized by the expression of cell surface markers, the relative expression of 6 genes, the secretion of 10 cytokines and the ability to form vessel-like structures. The cell proangiogenic properties was analysed in vivo, in a hindlimb ischemia model. Perfusion of lower limbs and functional tests were assessed for 28 days after cell infusion. Muscle histological analysis (neoangiogenesis, arteriogenesis and muscle repair) was performed.

Results: S-MSCs can be obtained from CLI-patients BMCs. They do not express endothelial specific markers but can be distinguished from MSCs by their secretome. S-MSCs have the ability to form tube-like structures and, in vivo, to induce blood flow recovery. No amputation was observed in S-MSCs treated mice. Functional tests showed improvement in treated groups with a superiority of MSCs and S-MSCs. In muscles, CD31+ and αSMA+ labelling were the highest in S-MSCs treated mice. S-MSCs induced the highest muscle repair.

Conclusions: S-MSCs exert angiogenic potential probably mediated by a paracrine mechanism. Their administration is associated with flow recovery, limb salvage and muscle repair. The secretome from S-MSCs or secretome-derived products may have a strong potential in vessel regeneration and muscle repair. Trial registration NCT00533104.

Keywords: Angiogenesis; Cell therapy; Critical limb ischemia; Mesenchymal stem cells.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Study design. a BMCs were obtained from 7 CLI patients. MSCs were selected and expanded in a CFU-F medium for 28 days (n = 7). Stimulated MSCs were cultured in EGM-2 medium for 14 days (n = 7). b Hindlimb ischemia was induced by femoral artery ligation in Nude mice. Cells (BMCs, MSCs, S-MSCs) or vehicle were injected in the gastrocnemius. Hindlimb perfusion, Necrose detection and functional tests were performed during 28 days. At day 28 muscles were harvested for biological analysis
Fig. 2
Fig. 2
Cells observation and real-time RT-PCR analysis. a BMCs morphology in May Grünwald Giemsa staining. b MSCs morphology in phase contrast microscopy. c S-MSCs morphology in phase contrast microscopy. d mRNA expression of six genes of interest were quantified using simultaneous real-time RT-qPCR experiments in MSCs (black box, n = 7), in S-MSCs (empty box, n = 7) and cb-ECFC (grey box, n = 5) used as control. x: aberrant distribution values as indicated by Box and Whiskers plots Medcalc version 7.3 software (*p < 0.05 and †p < 0.01)
Fig. 3
Fig. 3
Cells characterization by FC. a BMCs characterization. HSCs (CD34+ cells) analysis was performed by FC according to the ISHAGE reference method. b Immunophenotype characterization of isolated MSCs and S-MSCs. MSCs and S-MSCs (grey) were stained with the following human monoclonal antibodies: CD105 (endoglin), CD90 (Thy-1), CD73 (5′ nucleotidase), CD34, CD45, CD11b (integrin alpha M), HLA-DR (human leucocyte antigen-D related), CD146 (MCAM), CD140a (PDGF RA), CD140b (PDGF RB), CD31 (PECAM1), CD144 (Ve-Cadherin), VEGF R1, VEGF-R2, CD184 (CXCR4) and CD106 (VCAM1). The isotype-matched mouse IgG1-FITC, IgG1-PE, IgG1-PCy5 and IgG1- PCy7 were used as negative controls (empty)
Fig. 4
Fig. 4
Angiogenic effect of culture supernates. a MSCs and S-MSCs culture supernates and cell-free media (CFU-F and EGM-2) were recovered at day 28. HMEC-1 were suspended in endothelial cell growth medium MV (n = 3), in cell-free CFU-F medium (n = 3), in cell-free EGM-2 medium (n = 3), in MSCs culture supernates (obtained from 5 CLI-MSCs, n = 2 each group), in S-MSCs culture supernates (obtained from 5 CLI-S-MSCs, n = 2 each group) and incubated on Matrigel during 24 h. The extend of the network of the capillary-like tubes was appreciated at 3:30 h. b Quantification of the loops number at 3:30 h. c Quantification of total tube length (µm) at 3:30 h. x: aberrant distribution values as indicated by Box and Whiskers plots Medcalc version 7.3 software (*p < 0.05, †p < 0.01 and ‡p < 0.001)
Fig. 5
Fig. 5
Hindlimb blood flow recovery. a Representative LASCA images show improved perfusion in vehicle-, BMCs-, MSCs- and S-MSCs- treated mice at days 0 after ligation, 3 and 28. Low or no blood perfusion was displayed as dark blue, whereas the highest perfusion was displayed as red. b Hindlimb blood flow recovery after femoral artery ligation and BMCs (n = 40), MSCs (n = 35), S-MSCs (n = 40), or vehicle (n = 20) injection. CT by BMCs transplantation (blue) was effective in comparison with vehicle (green) injection for blood flow recovery at day 28 but BMCs did not restore completely the blood flow. In contrast MSCs (red) provided complete recovery at day 20 whereas S-MSCs (violet) were effective to completely restore blood flow at day 14. c Hindlimb blood flow recovery after BMCs (approximately 6 mice per patient), MSCs (approximately 6 mice per patient) and S-MSCs (approximately 6 mice per patient) infusion. BMCs were obtained from 7 different CLI-patients (Table 1) and were considered as “gold standard” (*comparison of MSCs group versus BMCs group; #S-MSCs versus BMCs; ¥S-MSCs versus MSCs; §BMCs versus vehicle; ΔMSCs versus vehicle; ∞S-MSCs versus vehicle)
Fig. 6
Fig. 6
Limb survival. a S-MSCs provide complete limb salvage in comparison with MSCs and BMCs. b In comparison with vehicle (n = 20), BMCs (n = 40) provided better limb salvage (p < 0.001). There was no difference between BMCs and MSCs (n = 35) treatment. S-MSCs (n = 40) were the most protective compared to BMCs and MSCs (p < 0.05)
Fig. 7
Fig. 7
Functional tests (static and dynamic). a, b Static tests: MSCs and S-MSCs were significantly more effective than BMCs and vehicle to improve IDS. MSCs (red) and S-MSCs (violet) provided significantly higher ratios at day 28 in comparison with BMCs (blue). There was no difference between the MSCs and S-MSCs groups. c Dynamic test: S-MSCs, MSCs and BMCs were effective to restore a normal walk in comparison with vehicle [*comparison of MSCs group (n = 35) versus BMCs group (n = 40); #S-MSCs (n = 40) versus BMCs; ¥S-MSCs versus MSCs; §BMCs versus vehicle (n = 20); ΔMSCs versus vehicle; ∞S-MSCs versus vehicle]
Fig. 8
Fig. 8
Neovessels visualization with CD31 and αSMA labelling. CD31 + staining (red) detects both angiogenic capillaries and arterioles. αSMA+ (green) detects arterioles. a Visualization of capillaries and arterioles in the semimembranosus in normal and ischemic legs. b Visualization of capillaries and arterioles in the gastrocnemius in normal and ischemic legs. c Angiogenesis analysis: fluorescence intensity ratio (CD31+ labelling) in the ischemic gastrocnemius in comparison with controlateral muscle. d Arteriogenesis analysis: Fluorescence intensity ratio (αSMA+/CD31+ labelling) in the ischemic gastrocnemius in comparison with controlateral muscle (*comparison of MSCs group versus BMCs group; #S-MSCs versus BMCs; ¥S-MSCs versus MSCs; §BMCs versus vehicle; ΔMSCs versus vehicle; ∞S-MSCs versus vehicle)
Fig. 9
Fig. 9
Evaluation of muscle repair. a Histological analysis of semimembranosus muscle in normal and ischemic leg. b Histological analysis of gastrocnemius muscle in normal and ischemic leg. c Evaluation of muscle repair: percentage of muscle fibers with a central nucleus quantified in the ischemic gastrocnemius in comparison with its controlateral muscle (*comparison of MSCs group versus BMCs group; #S-MSCs versus BMCs; ¥S-MSCs versus MSCs; §BMCs versus vehicle; ΔMSCs versus vehicle; ∞S-MSCs versus vehicle)

References

    1. Criqui MH, Aboyans V. Epidemiology of peripheral artery disease. Circ Res. 2015;116:1509–1526. doi: 10.1161/CIRCRESAHA.116.303849.
    1. Norgren L, Hiatt WR, Dormandy JA, Nehler MR, Harris KA, Fowkes FG. Inter-society consensus for the management of peripheral arterial disease (TASC II) J Vasc Surg. 2007;45(Suppl S):S5–S67. doi: 10.1016/j.jvs.2006.12.037.
    1. Gupta NK, Armstrong EJ, Parikh SA. The current state of stem cell therapy for peripheral artery disease. Curr Cardiol Rep. 2014;16:447. doi: 10.1007/s11886-013-0447-2.
    1. Tateishi-Yuyama E, Matsubara H, Murohara T, Ikeda U, Shintani S, Masaki H, Amano K, Kishimoto Y, Yoshimoto K, Akashi H, et al. Therapeutic angiogenesis for patients with limb ischaemia by autologous transplantation of bone-marrow cells: a pilot study and a randomised controlled trial. Lancet. 2002;360:427–435. doi: 10.1016/S0140-6736(02)09670-8.
    1. Ai M, Yan CF, Xia FC, Zhou SL, He J, Li CP. Safety and efficacy of cell-based therapy on critical limb ischemia: a meta-analysis. Cytotherapy. 2016;18:712–724. doi: 10.1016/j.jcyt.2016.02.009.
    1. Rigato M, Monami M, Fadini GP. Autologous cell therapy for peripheral arterial disease: systematic review and meta-analysis of randomized, nonrandomized, and noncontrolled studies. Circ Res. 2017;120:1326–1340. doi: 10.1161/CIRCRESAHA.116.309045.
    1. Peeters Weem SM, Teraa M, de Borst GJ, Verhaar MC, Moll FL. Bone marrow derived cell therapy in critical limb ischemia: a meta-analysis of randomized placebo controlled trials. Eur J Vasc Endovasc Surg. 2015;50:775–783. doi: 10.1016/j.ejvs.2015.08.018.
    1. Liew A, Bhattacharya V, Shaw J, Stansby G. Cell therapy for critical limb ischemia: a meta-analysis of randomized controlled trials. Angiology. 2016;67:444–455. doi: 10.1177/0003319715595172.
    1. Dimmeler S, Leri A. Aging and disease as modifiers of efficacy of cell therapy. Circ Res. 2008;102:1319–1330. doi: 10.1161/CIRCRESAHA.108.175943.
    1. Pignon B, Sevestre MA, Kanagaratnam L, Pernod G, Stephan D, Emmerich J, Clement C, Sarlon G, Boulon C, Tournois C, Nguyen P. Autologous bone marrow mononuclear cell implantation and its impact on the outcome of patients with critical limb ischemia—results of a randomized, double-blind, placebo-controlled trial. Circ J. 2017;81:1713–1720. doi: 10.1253/circj.CJ-17-0045.
    1. Tournois C, Pignon B, Sevestre MA, Al-Rifai R, Creuza V, Poitevin G, Francois C, Nguyen P. Cell therapy in critical limb ischemia: a comprehensive analysis of two cell therapy products. Cytotherapy. 2017;19:299–310. doi: 10.1016/j.jcyt.2016.10.013.
    1. Friedenstein AJ, Gorskaja JF, Kulagina NN. Fibroblast precursors in normal and irradiated mouse hematopoietic organs. Exp Hematol. 1976;4:267–274.
    1. Mushahary D, Spittler A, Kasper C, Weber V, Charwat V. Isolation, cultivation, and characterization of human mesenchymal stem cells. Cytom A. 2018;93:19–31. doi: 10.1002/cyto.a.23242.
    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. Vizoso FJ, Eiro N, Cid S, Schneider J, Perez-Fernandez R. Mesenchymal stem cell secretome: toward cell-free therapeutic strategies in regenerative medicine. Int J Mol Sci. 2017;18:1852–76. doi: 10.3390/ijms18091852.
    1. Yong KW, Choi JR, Mohammadi M, Mitha AP, Sanati-Nezhad A, Sen A. Mesenchymal stem cell therapy for ischemic tissues. Stem Cells Int. 2018;2018:8179075. doi: 10.1155/2018/8179075.
    1. Lu D, Chen B, Liang Z, Deng W, Jiang Y, Li S, Xu J, Wu Q, Zhang Z, Xie B, Chen S. Comparison of bone marrow mesenchymal stem cells with bone marrow-derived mononuclear cells for treatment of diabetic critical limb ischemia and foot ulcer: a double-blind, randomized, controlled trial. Diabetes Res Clin Pract. 2011;92:26–36. doi: 10.1016/j.diabres.2010.12.010.
    1. Gupta PK, Chullikana A, Parakh R, Desai S, Das A, Gottipamula S, Krishnamurthy S, Anthony N, Pherwani A, Majumdar AS. A double blind randomized placebo controlled phase I/II study assessing the safety and efficacy of allogeneic bone marrow derived mesenchymal stem cell in critical limb ischemia. J Transl Med. 2013;11:143. doi: 10.1186/1479-5876-11-143.
    1. Bura A, Planat-Benard V, Bourin P, Silvestre JS, Gross F, Grolleau JL, Saint-Lebese B, Peyrafitte JA, Fleury S, Gadelorge M, et al. Phase I trial: the use of autologous cultured adipose-derived stroma/stem cells to treat patients with non-revascularizable critical limb ischemia. Cytotherapy. 2014;16:245–257. doi: 10.1016/j.jcyt.2013.11.011.
    1. Barreto-Duran E, Mejia-Cruz CC, Leal-Garcia E, Perez-Nunez R, Rodriguez-Pardo VM. Impact of donor characteristics on the quality of bone marrow as a source of mesenchymal stromal cells. Am J Stem Cells. 2018;7:114–120.
    1. Liu JW, Dunoyer-Geindre S, Serre-Beinier V, Mai G, Lambert JF, Fish RJ, Pernod G, Buehler L, Bounameaux H, Kruithof EK. Characterization of endothelial-like cells derived from human mesenchymal stem cells. J Thromb Haemost. 2007;5:826–834. doi: 10.1111/j.1538-7836.2007.02381.x.
    1. Ning H, Liu G, Lin G, Yang R, Lue TF, Lin CS. Fibroblast growth factor 2 promotes endothelial differentiation of adipose tissue-derived stem cells. J Sex Med. 2009;6:967–979. doi: 10.1111/j.1743-6109.2008.01172.x.
    1. Konno M, Hamazaki TS, Fukuda S, Tokuhara M, Uchiyama H, Okazawa H, Okochi H, Asashima M. Efficiently differentiating vascular endothelial cells from adipose tissue-derived mesenchymal stem cells in serum-free culture. Biochem Biophys Res Commun. 2010;400:461–465. doi: 10.1016/j.bbrc.2010.08.029.
    1. Bayes-Genis A, Soler-Botija C, Farre J, Sepulveda P, Raya A, Roura S, Prat-Vidal C, Galvez-Monton C, Montero JA, Buscher D, Izpisua Belmonte JC. Human progenitor cells derived from cardiac adipose tissue ameliorate myocardial infarction in rodents. J Mol Cell Cardiol. 2010;49:771–780. doi: 10.1016/j.yjmcc.2010.08.010.
    1. Roura S, Bago JR, Soler-Botija C, Pujal JM, 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. Gaafar TM, Abdel Rahman HA, Attia W, Hamza HS, Brockmeier K, El Hawary RE. Comparative characteristics of endothelial-like cells derived from human adipose mesenchymal stem cells and umbilical cord blood-derived endothelial cells. Clin Exp Med. 2014;14:177–184. doi: 10.1007/s10238-013-0238-5.
    1. Cannella V, Piccione G, Altomare R, Marino A, Di Marco P, Russotto L, Di Bella S, Purpari G, Gucciardi F, Cassata G, et al. Differentiation and characterization of rat adipose tissue mesenchymal stem cells into endothelial-like cells. Anat Histol Embryol. 2018;47:11–20. doi: 10.1111/ahe.12318.
    1. Cuccuini W, Poitevin S, Poitevin G, Dignat-George F, Cornillet-Lefebvre P, Sabatier F, Nguyen P. Tissue factor up-regulation in proinflammatory conditions confers thrombin generation capacity to endothelial colony-forming cells without influencing non-coagulant properties in vitro. J Thromb Haemost. 2010;8:2042–2052. doi: 10.1111/j.1538-7836.2010.03936.x.
    1. Gremmels H, Teraa M, Quax PH, den Ouden K, Fledderus JO, Verhaar MC. Neovascularization capacity of mesenchymal stromal cells from critical limb ischemia patients is equivalent to healthy controls. Mol Ther. 2014;22:1960–1970. doi: 10.1038/mt.2014.161.
    1. Limbourg A, Korff T, Napp LC, Schaper W, Drexler H, Limbourg FP. Evaluation of postnatal arteriogenesis and angiogenesis in a mouse model of hind-limb ischemia. Nat Protoc. 2009;4:1737–1746. doi: 10.1038/nprot.2009.185.
    1. Brenes RA, Jadlowiec CC, Bear M, Hashim P, Protack CD, Li X, Lv W, Collins MJ, Dardik A. Toward a mouse model of hind limb ischemia to test therapeutic angiogenesis. J Vasc Surg. 2012;56:1669–1679. doi: 10.1016/j.jvs.2012.04.067.
    1. Choi M, Lee HS, Naidansaren P, Kim HK, Eunju O, Cha JH, Ahn HY, Yang PI, Shin JC, Joe YA. Proangiogenic features of Wharton’s jelly-derived mesenchymal stromal/stem cells and their ability to form functional vessels. Int J Biochem Cell Biol. 2013;45:560–570. doi: 10.1016/j.biocel.2012.12.001.
    1. Reis PE, de Carvalho LP, Yasumura E, da Silva FH, Garcia BC, Beutel A, Sacramento CB, Baptista-Silva JC, de Campos RR, Takiya CM, et al. Impact of angiogenic therapy in the treatment of critical lower limb ischemia in an animal model. Vasc Endovasc Surg. 2014;48:207–216. doi: 10.1177/1538574413518119.
    1. Cadot B, Gache V, Gomes ER. Moving and positioning the nucleus in skeletal muscle—one step at a time. Nucleus. 2015;6:373–381. doi: 10.1080/19491034.2015.1090073.
    1. Roman W, Gomes ER. Nuclear positioning in skeletal muscle. Semin Cell Dev Biol. 2018;82:51–56. doi: 10.1016/j.semcdb.2017.11.005.
    1. Smadja DM, d’Audigier C, Guerin CL, Mauge L, Dizier B, Silvestre JS, Dal Cortivo L, Gaussem P, Emmerich J. Angiogenic potential of BM MSCs derived from patients with critical leg ischemia. Bone Marrow Transplant. 2012;47:997–1000. doi: 10.1038/bmt.2011.196.
    1. Wang C, Li Y, Yang M, Zou Y, Liu H, Liang Z, Yin Y, Niu G, Yan Z, Zhang B. Efficient differentiation of bone marrow mesenchymal stem cells into endothelial cells in vitro. Eur J Vasc Endovasc Surg. 2018;55:257–265. doi: 10.1016/j.ejvs.2017.10.012.
    1. Oswald J, Boxberger S, Jorgensen B, Feldmann S, Ehninger G, Bornhauser M, Werner C. Mesenchymal stem cells can be differentiated into endothelial cells in vitro. Stem Cells. 2004;22:377–384. doi: 10.1634/stemcells.22-3-377.
    1. Tancharoen W, Aungsuchawan S, Pothacharoen P, Markmee R, Narakornsak S, Kieodee J, Boonma N, Tasuya W. Differentiation of mesenchymal stem cells from human amniotic fluid to vascular endothelial cells. Acta Histochem. 2017;119:113–121. doi: 10.1016/j.acthis.2016.11.009.
    1. Janeczek Portalska K, Leferink A, Groen N, Fernandes H, Moroni L, van Blitterswijk C, de Boer J. Endothelial differentiation of mesenchymal stromal cells. PLoS ONE. 2012;7:e46842. doi: 10.1371/journal.pone.0046842.
    1. Policha A, Zhang P, Chang L, Lamb K, Tulenko T, DiMuzio P. Endothelial differentiation of diabetic adipose-derived stem cells. J Surg Res. 2014;192:656–663. doi: 10.1016/j.jss.2014.06.041.
    1. Chen L, Xu Y, Zhao J, Zhang Z, Yang R, Xie J, Liu X, Qi S. Conditioned medium from hypoxic bone marrow-derived mesenchymal stem cells enhances wound healing in mice. PLoS ONE. 2014;9:e96161. doi: 10.1371/journal.pone.0096161.
    1. Teo GS, Ankrum JA, Martinelli R, Boetto SE, Simms K, Sciuto TE, Dvorak AM, Karp JM, Carman CV. Mesenchymal stem cells transmigrate between and directly through tumor necrosis factor-alpha-activated endothelial cells via both leukocyte-like and novel mechanisms. Stem Cells. 2012;30:2472–2486. doi: 10.1002/stem.1198.
    1. Belmadani S, Matrougui K, Kolz C, Pung YF, Palen D, Prockop DJ, Chilian WM. Amplification of coronary arteriogenic capacity of multipotent stromal cells by epidermal growth factor. Arterioscler Thromb Vasc Biol. 2009;29:802–808. doi: 10.1161/ATVBAHA.109.186189.
    1. Goedecke A, Wobus M, Krech M, Munch N, Richter K, Holig K, Bornhauser M. Differential effect of platelet-rich plasma and fetal calf serum on bone marrow-derived human mesenchymal stromal cells expanded in vitro. J Tissue Eng Regen Med. 2011;5:648–654. doi: 10.1002/term.359.
    1. Harris LJ, Zhang P, Abdollahi H, Tarola NA, DiMatteo C, McIlhenny SE, Tulenko TN, DiMuzio PJ. Availability of adipose-derived stem cells in patients undergoing vascular surgical procedures. J Surg Res. 2010;163:e105–e112. doi: 10.1016/j.jss.2010.04.025.
    1. Simons M, Alitalo K, Annex BH, Augustin HG, Beam C, Berk BC, Byzova T, Carmeliet P, Chilian W, Cooke JP, et al. State-of-the-art methods for evaluation of angiogenesis and tissue vascularization: a scientific statement from the American Heart Association. Circ Res. 2015;116:e99–e132. doi: 10.1161/CIRCRESAHA.116.304710.
    1. Iwase T, Nagaya N, Fujii T, Itoh T, Murakami S, Matsumoto T, Kangawa K, Kitamura S. Comparison of angiogenic potency between mesenchymal stem cells and mononuclear cells in a rat model of hindlimb ischemia. Cardiovasc Res. 2005;66:543–551. doi: 10.1016/j.cardiores.2005.02.006.
    1. Morris AD, Dalal S, Li H, Brewster LP. Human diabetic mesenchymal stem cells from peripheral arterial disease patients promote angiogenesis through unique secretome signatures. Surgery. 2018;163:870–876. doi: 10.1016/j.surg.2017.11.018.
    1. Brewster L, Robinson S, Wang R, Griffiths S, Li H, Peister A, Copland I, McDevitt T. Expansion and angiogenic potential of mesenchymal stem cells from patients with critical limb ischemia. J Vasc Surg. 2017;65(826–838):e821.
    1. Bergers G, Song S. The role of pericytes in blood-vessel formation and maintenance. Neuro Oncol. 2005;7:452–464. doi: 10.1215/S1152851705000232.
    1. van Royen N, Piek JJ, Schaper W, Fulton WF. A critical review of clinical arteriogenesis research. J Am Coll Cardiol. 2009;55:17–25. doi: 10.1016/j.jacc.2009.06.058.
    1. Hakimzadeh N, Verberne HJ, Siebes M, Piek JJ. The future of collateral artery research. Curr Cardiol Rev. 2014;10:73–86. doi: 10.2174/1573403X113099990001.
    1. Hendrikx G, Voo S, Bauwens M, Post MJ, Mottaghy FM. SPECT and PET imaging of angiogenesis and arteriogenesis in pre-clinical models of myocardial ischemia and peripheral vascular disease. Eur J Nucl Med Mol Imaging. 2016;43:2433–2447. doi: 10.1007/s00259-016-3480-8.
    1. Beauchamp JR, Morgan JE, Pagel CN, Partridge TA. Dynamics of myoblast transplantation reveal a discrete minority of precursors with stem cell-like properties as the myogenic source. J Cell Biol. 1999;144:1113–1122. doi: 10.1083/jcb.144.6.1113.
    1. Lee RH, Pulin AA, Seo MJ, Kota DJ, Ylostalo J, Larson BL, Semprun-Prieto L, Delafontaine P, Prockop DJ. Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. Cell Stem Cell. 2009;5:54–63. doi: 10.1016/j.stem.2009.05.003.
    1. Koutakis P, Myers SA, Cluff K, Ha DM, Haynatzki G, McComb RD, Uchida K, Miserlis D, Papoutsi E, Johanning JM, et al. Abnormal myofiber morphology and limb dysfunction in claudication. J Surg Res. 2015;196:172–179. doi: 10.1016/j.jss.2015.02.011.
    1. Heemskerk AM, Strijkers GJ, Drost MR, van Bochove GS, Nicolay K. Skeletal muscle degeneration and regeneration after femoral artery ligation in mice: monitoring with diffusion MR imaging. Radiology. 2007;243:413–421. doi: 10.1148/radiol.2432060491.
    1. Kim SW, Han H, Chae GT, Lee SH, Bo S, Yoon JH, Lee YS, Lee KS, Park HK, Kang KS. Successful stem cell therapy using umbilical cord blood-derived multipotent stem cells for Buerger’s disease and ischemic limb disease animal model. Stem Cells. 2006;24:1620–1626. doi: 10.1634/stemcells.2005-0365.
    1. Dash NR, Dash SN, Routray P, Mohapatra S, Mohapatra PC. Targeting nonhealing ulcers of lower extremity in human through autologous bone marrow-derived mesenchymal stem cells. Rejuvenation Res. 2009;12:359–366. doi: 10.1089/rej.2009.0872.
    1. Lasala GP, Silva JA, Gardner PA, Minguell JJ. Combination stem cell therapy for the treatment of severe limb ischemia: safety and efficacy analysis. Angiology. 2010;61:551–556. doi: 10.1177/0003319710364213.
    1. Powell RJ, Marston WA, Berceli SA, Guzman R, Henry TD, Longcore AT, Stern TP, Watling S, Bartel RL. Cellular therapy with Ixmyelocel-T to treat critical limb ischemia: the randomized, double-blind, placebo-controlled RESTORE-CLI trial. Mol Ther. 2012;20:1280–1286. doi: 10.1038/mt.2012.52.
    1. Lasala GP, Silva JA, Minguell JJ. Therapeutic angiogenesis in patients with severe limb ischemia by transplantation of a combination stem cell product. J Thorac Cardiovasc Surg. 2012;144:377–382. doi: 10.1016/j.jtcvs.2011.08.053.
    1. Lee HC, An SG, Lee HW, Park JS, Cha KS, Hong TJ, Park JH, Lee SY, Kim SP, Kim YD, et al. Safety and effect of adipose tissue-derived stem cell implantation in patients with critical limb ischemia: a pilot study. Circ J. 2012;76:1750–1760. doi: 10.1253/circj.CJ-11-1135.
    1. Das AK, Bin Abdullah BJ, Dhillon SS, Vijanari A, Anoop CH, Gupta PK. Intra-arterial allogeneic mesenchymal stem cells for critical limb ischemia are safe and efficacious: report of a phase I study. World J Surg. 2013;37:915–922. doi: 10.1007/s00268-012-1892-6.
    1. Yang SS, Kim NR, Park KB, Do YS, Roh K, Kang KS, Kim DI. A phase I study of human cord blood-derived mesenchymal stem cell therapy in patients with peripheral arterial occlusive disease. Int J Stem Cells. 2013;6:37–44. doi: 10.15283/ijsc.2013.6.1.37.
    1. Powell RJ, Comerota AJ, Berceli SA, Guzman R, Henry TD, Tzeng E, Velazquez O, Marston WA, Bartel RL, Longcore A, et al. Interim analysis results from the RESTORE-CLI, a randomized, double-blind multicenter phase II trial comparing expanded autologous bone marrow-derived tissue repair cells and placebo in patients with critical limb ischemia. J Vasc Surg. 2011;54:1032–1041. doi: 10.1016/j.jvs.2011.04.006.
    1. El Omar R, Xiong Y, Dostert G, Louis H, Gentils M, Menu P, Stoltz JF, Velot E, Decot V. Immunomodulation of endothelial differentiated mesenchymal stromal cells: impact on T and NK cells. Immunol Cell Biol. 2016;94:342–356. doi: 10.1038/icb.2015.94.
    1. Seo Y, Kim HS, Hong IS. Stem cell-derived extracellular vesicles as immunomodulatory therapeutics. Stem Cells Int. 2019;2019:5126156.
    1. Phelps J, Sanati-Nezhad A, Ungrin M, Duncan NA, Sen A. Bioprocessing of mesenchymal stem cells and their derivatives: toward cell-free therapeutics. Stem Cells Int. 2018;2018:9415367. doi: 10.1155/2018/9415367.

Source: PubMed

3
Abonnieren