Skip to main content

Introduction to Stem Cell Therapy and Its Application in Vascular Diseases

  • Chapter
  • First Online:
Stem Cell Therapy for Vascular Diseases

Abstract

Stem cell therapy has recently emerged as an innovative therapeutic strategy for various vascular diseases. Stem cell therapy comprises a spectrum of cell-based regenerative strategies characterized by three sets of variables: (1) stem cell intrinsic properties (origin, type, isolation/induction, culture, and priming features); (2) therapy protocol (dose, concentration, administration route, adjuvants); and (3) patient characteristics (medical history, immune reaction, disease of interest). In this chapter, we introduce basic concepts regarding stem cell therapy: stem cell types and origin, mechanisms of regeneration, isolation, cell culture and priming, administration routes, and therapeutic risks. We also discuss the effects and interactions between these variables as well as their impact in the treatment of vascular diseases.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 84.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 109.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 109.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Zakrzewski W, Dobrzyński M, Szymonowicz M, Rybak Z. Stem cells: past, present, and future. Stem Cell Res Ther. 2019;10(1):68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Kirby GTS, Mills SJ, Cowin AJ, Smith LE. Stem cells for cutaneous wound healing. Biomed Res Int. 2015;2015:285869.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Konstantinov IE. In search of Alexander a. Maximow: the man behind the unitarian theory of hematopoiesis. Perspect Biol Med. 2000;43(2):269–76.

    Article  CAS  PubMed  Google Scholar 

  4. Till JE, McCulloch EA. A direct measurement of the radiation sensitivity of normal mouse bone marrow cells. Radiat Res. 1961;14(2):213–22.

    Article  CAS  PubMed  Google Scholar 

  5. Gurdon JB. The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles. J Embryol Exp Morphol. 1962;10:622–40.

    CAS  PubMed  Google Scholar 

  6. Friedenstein AJ, Piatetzky S II, Petrakova KV. Osteogenesis in transplants of bone marrow cells. J Embryol Exp Morphol. 1966;16(3):381–90.

    CAS  PubMed  Google Scholar 

  7. de la Morena MT, Gatti RA. A history of bone marrow transplantation. Hematol Oncol Clin North Am. 2011;25(1):1–15.

    Article  PubMed  Google Scholar 

  8. Evans MJ, Kaufman MH. Establishment in culture of pluripotential cells from mouse embryos. Nature. 1981;292(5819):154–6.

    Article  CAS  PubMed  Google Scholar 

  9. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282(5391):1145–7.

    Article  CAS  PubMed  Google Scholar 

  10. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–76.

    Article  CAS  PubMed  Google Scholar 

  11. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861–72.

    Article  CAS  PubMed  Google Scholar 

  12. NobelPrize.org. The Nobel prize in physiology or medicine 2012. [cited 2020 04 April]; Available from: https://www.nobelprize.org/prizes/medicine/2012/press-release/.

  13. Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991;9(5):641–50.

    Article  CAS  PubMed  Google Scholar 

  14. Yin JQ, Zhu J, Ankrum JA. Manufacturing of primed mesenchymal stromal cells for therapy. Nat Biomed Eng. 2019;3(2):90–104.

    Article  CAS  PubMed  Google Scholar 

  15. Horwitz EM, Le Blanc K, Dominici M, Mueller I, Slaper-Cortenbach I, Marini FC, et al. Clarification of the nomenclature for MSC: the International Society for Cellular Therapy position statement. Cytotherapy. 2005;7(5):393–5.

    Article  CAS  PubMed  Google Scholar 

  16. Viswanathan S, Shi Y, Galipeau J, Krampera M, Leblanc K, Martin I, et al. Mesenchymal stem versus stromal cells: International Society for Cell & Gene Therapy (ISCT(R)) mesenchymal stromal cell committee position statement on nomenclature. Cytotherapy. 2019;21(10):1019–24.

    Article  CAS  PubMed  Google Scholar 

  17. Lin CS, Ning H, Lin G, Lue TF. Is CD34 truly a negative marker for mesenchymal stromal cells? Cytotherapy. 2012;14(10):1159–63.

    Article  CAS  PubMed  Google Scholar 

  18. Bellagamba BC, Grudzinski PB, Ely PB, Nader PJH, Nardi NB, da Silva Meirelles L. Induction of expression of CD271 and CD34 in mesenchymal stromal cells cultured as spheroids. Stem Cells Int. 2018;2018:7357213.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Available from: https://clinicaltrials.gov/ct2/results?cond=&term=mesenchymal+stem+cell&cntry=&state=&city=&dist=.

  20. Leeper NJ, Hunter AL, Cooke JP. Stem cell therapy for vascular regeneration: adult, embryonic, and induced pluripotent stem cells. Circulation. 2010;122(5):517–26.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Blum B, Benvenisty N. The tumorigenicity of human embryonic stem cells. Adv Cancer Res. 2008;100:133–58.

    Article  PubMed  Google Scholar 

  22. Mali P, Ye Z, Hommond HH, Yu X, Lin J, Chen G, et al. Improved efficiency and pace of generating induced pluripotent stem cells from human adult and fetal fibroblasts. Stem Cells (Dayton, Ohio). 2008;26(8):1998–2005.

    Article  CAS  Google Scholar 

  23. Choudhery MS, Badowski M, Muise A, Pierce J, Harris DT. Donor age negatively impacts adipose tissue-derived mesenchymal stem cell expansion and differentiation. J Transl Med. 2014;12:8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Beane OS, Fonseca VC, Cooper LL, Koren G, Darling EM. Impact of aging on the regenerative properties of bone marrow-, muscle-, and adipose-derived mesenchymal stem/stromal cells. PLoS One. 2014;9(12):e115963.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Duscher D, Rennert RC, Januszyk M, Anghel E, Maan ZN, Whittam AJ, et al. Aging disrupts cell subpopulation dynamics and diminishes the function of mesenchymal stem cells. Sci Rep. 2014;4:7144.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hequet O. Hematopoietic stem and progenitor cell harvesting: technical advances and clinical utility. J Blood Med. 2015;6:55–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Serena C, Keiran N, Ceperuelo-Mallafre V, Ejarque M, Fradera R, Roche K, et al. Obesity and type 2 diabetes alters the immune properties of human adipose derived stem cells. Stem Cells. 2016;34(10):2559–73.

    Article  CAS  PubMed  Google Scholar 

  28. Berglund AK, Fortier LA, Antczak DF, Schnabel LV. Immunoprivileged no more: measuring the immunogenicity of allogeneic adult mesenchymal stem cells. Stem Cell Res Ther. 2017;8(1):288.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Galleu A, Riffo-Vasquez Y, Trento C, Lomas C, Dolcetti L, Cheung TS, et al. Apoptosis in mesenchymal stromal cells induces in vivo recipient-mediated immunomodulation. Sci Transl Med. 2017;9(416):3–4.

    Google Scholar 

  30. Hodgetts SI, Stagg K, Sturm M, Edel M, Blancafort P. Long live the stem cell: the use of stem cells isolated from post mortem tissues for translational strategies. Int J Biochem Cell Biol. 2014;56:74–81.

    Article  CAS  PubMed  Google Scholar 

  31. Administration FUSFD. Xenotransplantation. 2017 [updated 04/28/2017; cited 2017 12/05/2017]; Available from: https://www.fda.gov/BiologicsBloodVaccines/Xenotransplantation/.

  32. Hass R, Kasper C, Bohm S, Jacobs R. Different populations and sources of human mesenchymal stem cells (MSC): a comparison of adult and neonatal tissue-derived MSC. Cell Commun Signal. 2011;9:12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Murray IR, West CC, Hardy WR, James AW, Park TS, Nguyen A, et al. Natural history of mesenchymal stem cells, from vessel walls to culture vessels. Cell Mol Life Sci. 2014;71(8):1353–74.

    Article  CAS  PubMed  Google Scholar 

  34. Andia I, Maffulli N, Burgos-Alonso N. Stromal vascular fraction technologies and clinical applications. Expert Opin Biol Ther. 2019;19(12):1289–305.

    Article  PubMed  Google Scholar 

  35. Gentile P, Calabrese C, De Angelis B, Pizzicannella J, Kothari A, Garcovich S. Impact of the different preparation methods to obtain human adipose-derived stromal vascular fraction cells (AD-SVFs) and human adipose-derived mesenchymal stem cells (AD-MSCs): enzymatic digestion versus mechanical centrifugation. Int J Mol Sci. 2019;20(21):4–7.

    Google Scholar 

  36. Lopes L, Setia O, Aurshina A, Liu S, Hu H, Isaji T, et al. Stem cell therapy for diabetic foot ulcers: a review of preclinical and clinical research. Stem Cell Res Ther. 2018;9(1):188.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Khan FA, Almohazey D, Alomari M, Almofty SA. Isolation, culture, and functional characterization of human embryonic stem cells: current trends and challenges. Stem Cells Int. 2018;2018:1429351.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Tanaka N, Takeuchi T, Neri QV, Sills ES, Palermo GD. Laser-assisted blastocyst dissection and subsequent cultivation of embryonic stem cells in a serum/cell free culture system: applications and preliminary results in a murine model. J Transl Med. 2006;4:20.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Strom S, Inzunza J, Grinnemo KH, Holmberg K, Matilainen E, Stromberg AM, et al. Mechanical isolation of the inner cell mass is effective in derivation of new human embryonic stem cell lines. Hum Reprod. 2007;22(12):3051–8.

    Article  PubMed  Google Scholar 

  40. Hayes M, Zavazava N. Strategies to generate induced pluripotent stem cells. Methods Mol Biol. 2013;1029:77–92.

    Article  CAS  PubMed  Google Scholar 

  41. Yeo JC, Ng HH. The transcriptional regulation of pluripotency. Cell Res. 2013;23(1):20–32.

    Article  CAS  PubMed  Google Scholar 

  42. Zhang S, Cui W. Sox2, a key factor in the regulation of pluripotency and neural differentiation. World J Stem Cells. 2014;6(3):305–11.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Dakhore S, Nayer B, Hasegawa K. Human pluripotent stem cell culture: current status, challenges, and advancement. Stem Cells Int. 2018;2018:7396905.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Gstraunthaler G, Lindl T, van der Valk J. A plea to reduce or replace fetal bovine serum in cell culture media. Cytotechnology. 2013;65(5):791–3.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Burnouf T, Strunk D, Koh MB, Schallmoser K. Human platelet lysate: replacing fetal bovine serum as a gold standard for human cell propagation? Biomaterials. 2016;76:371–87.

    Article  CAS  PubMed  Google Scholar 

  46. Abdelrazik H, Spaggiari GM, Chiossone L, Moretta L. Mesenchymal stem cells expanded in human platelet lysate display a decreased inhibitory capacity on T- and NK-cell proliferation and function. Eur J Immunol. 2011;41(11):3281–90.

    Article  CAS  PubMed  Google Scholar 

  47. Oikonomopoulos A, van Deen WK, Manansala AR, Lacey PN, Tomakili TA, Ziman A, et al. Optimization of human mesenchymal stem cell manufacturing: the effects of animal/xeno-free media. Sci Rep. 2015;5:16570.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Ejtehadifar M, Shamsasenjan K, Movassaghpour A, Akbarzadehlaleh P, Dehdilani N, Abbasi P, et al. The effect of hypoxia on mesenchymal stem cell biology. Adv Pharmaceut Bullet. 2015;5(2):141–9.

    Article  CAS  Google Scholar 

  49. Nold P, Hackstein H, Riedlinger T, Kasper C, Neumann A, Mernberger M, et al. Immunosuppressive capabilities of mesenchymal stromal cells are maintained under hypoxic growth conditions and after gamma irradiation. Cytotherapy. 2015;17(2):152–62.

    Article  CAS  PubMed  Google Scholar 

  50. Rosova I, Dao M, Capoccia B, Link D, Nolta JA. Hypoxic preconditioning results in increased motility and improved therapeutic potential of human mesenchymal stem cells. Stem Cells. 2008;26(8):2173–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Das B, Bayat-Mokhtari R, Tsui M, Lotfi S, Tsuchida R, Felsher DW, et al. HIF-2alpha suppresses p53 to enhance the stemness and regenerative potential of human embryonic stem cells. Stem Cells. 2012;30(8):1685–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Grayson WL, Zhao F, Izadpanah R, Bunnell B, Ma T. Effects of hypoxia on human mesenchymal stem cell expansion and plasticity in 3D constructs. J Cell Physiol. 2006;207(2):331–9.

    Article  CAS  PubMed  Google Scholar 

  53. Basciano L, Nemos C, Foliguet B, de Isla N, de Carvalho M, Tran N, et al. Long term culture of mesenchymal stem cells in hypoxia promotes a genetic program maintaining their undifferentiated and multipotent status. BMC Cell Biol. 2011;12:12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Roobrouck VD, Vanuytsel K, Verfaillie CM. Concise review: culture mediated changes in fate and/or potency of stem cells. Stem Cells. 2011;29(4):583–9.

    Article  CAS  PubMed  Google Scholar 

  55. Shekaran A, Sim E, Tan KY, Chan JKY, Choolani M, Reuveny S, et al. Enhanced in vitro osteogenic differentiation of human fetal MSCs attached to 3D microcarriers versus harvested from 2D monolayers. BMC Biotechnol. 2015;15(1):102.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Haugh MG, Heilshorn SC. Integrating concepts of material mechanics, ligand chemistry, dimensionality and degradation to control differentiation of mesenchymal stem cells. Curr Opin Solid State Mater Sci. 2016;20(4):171–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Mo M, Zhou Y, Li S, Wu Y. Three-dimensional culture reduces cell size by increasing vesicle excretion. Stem Cells. 2018;36(2):286–92.

    Article  CAS  PubMed  Google Scholar 

  58. Petrenko Y, Sykova E, Kubinova S. The therapeutic potential of three-dimensional multipotent mesenchymal stromal cell spheroids. Stem Cell Res Ther. 2017;8(1):94.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Rashedi I, Talele N, Wang XH, Hinz B, Radisic M, Keating A. Collagen scaffold enhances the regenerative properties of mesenchymal stromal cells. PLoS One. 2017;12(10):e0187348.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Murphy WL, McDevitt TC, Engler AJ. Materials as stem cell regulators. Nat Mater. 2014;13(6):547–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell. 2006;126(4):677–89.

    Article  CAS  PubMed  Google Scholar 

  62. Shakouri-Motlagh A, O'Connor AJ, Brennecke SP, Kalionis B, Heath DE. Native and solubilized decellularized extracellular matrix: a critical assessment of their potential for improving the expansion of mesenchymal stem cells. Acta Biomater. 2017;55:1–12.

    Article  CAS  PubMed  Google Scholar 

  63. Carrier-Ruiz A, Evaristo-Mendonca F, Mendez-Otero R, Ribeiro-Resende VT. Biological behavior of mesenchymal stem cells on poly-epsilon-caprolactone filaments and a strategy for tissue engineering of segments of the peripheral nerves. Stem Cell Res Ther. 2015;6:128.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Lam A, Li J, Toh J, Sim E, Chen A, Chan J, et al. Biodegradable poly-ε-caprolactone microcarriers for efficient production of human mesenchymal stromal cells and secreted cytokines in batch and fed-batch bioreactors. Cytotherapy. 2016;19:8–12.

    Google Scholar 

  65. Jun I, Lee YB, Choi YS, Engler AJ, Park H, Shin H. Transfer stamping of human mesenchymal stem cell patches using thermally expandable hydrogels with tunable cell-adhesive properties. Biomaterials. 2015;54:44–54.

    Article  CAS  PubMed  Google Scholar 

  66. Duscher D, Barrera J, Wong VW, Maan ZN, Whittam AJ, Januszyk M, et al. Stem cells in wound healing: the future of regenerative medicine? A mini-review. Gerontology. 2016;62(2):216–25.

    Article  CAS  PubMed  Google Scholar 

  67. Isakson M, de Blacam C, Whelan D, McArdle A, Clover AJP. Mesenchymal stem cells and cutaneous wound healing: current evidence and future potential. Stem Cells Int. 2015;2015:831095.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Caplan H, Olson SD, Kumar A, George M, Prabhakara KS, Wenzel P, et al. Mesenchymal stromal cell therapeutic delivery: translational challenges to clinical application. Front Immunol. 2019;10:1645.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. O’Loughlin A, O’Brien T. Topical stem and progenitor cell therapy for diabetic foot ulcers. In: Gholamrezanezhad DA, editor. Stem cells clinic and research. Published online: InTech; 2011. p. 578–604.

    Google Scholar 

  70. Hamidian Jahromi S, Davies JE. Concise review: skeletal muscle as a delivery route for mesenchymal stromal cells. Stem Cells Transl Med. 2019;8(5):456–65.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Qazi TH, Mooney DJ, Duda GN, Geissler S. Biomaterials that promote cell-cell interactions enhance the paracrine function of MSCs. Biomaterials. 2017;140:103–14.

    Article  CAS  PubMed  Google Scholar 

  72. Jenkins TL, Little D. Synthetic scaffolds for musculoskeletal tissue engineering: cellular responses to fiber parameters. NPJ Regen Med. 2019;4:15.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Falanga V, Iwamoto S, Chartier M, Yufit T, Butmarc J, Kouttab N, et al. Autologous bone marrow-derived cultured mesenchymal stem cells delivered in a fibrin spray accelerate healing in murine and human cutaneous wounds. Tissue Eng. 2007;13(6):1299–312.

    Article  CAS  PubMed  Google Scholar 

  74. Watanabe M, Yavagal DR. Intra-arterial delivery of mesenchymal stem cells. Brain Circ. 2016;2(3):114–7.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Janowski M, Lyczek A, Engels C, Xu J, Lukomska B, Bulte JW, et al. Cell size and velocity of injection are major determinants of the safety of intracarotid stem cell transplantation. J Cereb Blood Flow Metab. 2013;33(6):921–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Fischer UM, Harting MT, Jimenez F, Monzon-Posadas WO, Xue H, Savitz SI, et al. Pulmonary passage is a major obstacle for intravenous stem cell delivery: the pulmonary first-pass effect. Stem Cells Dev. 2009;18(5):683–92.

    Article  CAS  PubMed  Google Scholar 

  77. de Witte SFH, Luk F, Sierra Parraga JM, Gargesha M, Merino A, Korevaar SS, et al. Immunomodulation by therapeutic Mesenchymal Stromal Cells (MSC) is triggered through phagocytosis of MSC by Monocytic cells. Stem Cells. 2018;36(4):602–15.

    Article  PubMed  CAS  Google Scholar 

  78. Nijboer CH, Kooijman E, van Velthoven CT, van Tilborg E, Tiebosch IA, Eijkelkamp N, et al. Intranasal stem cell treatment as a novel therapy for subarachnoid hemorrhage. Stem Cells Dev. 2018;27(5):313–25.

    Article  CAS  PubMed  Google Scholar 

  79. Vaquero J, Zurita M, Rico MA, Aguayo C, Bonilla C, Marin E, et al. Intrathecal administration of autologous mesenchymal stromal cells for spinal cord injury: safety and efficacy of the 100/3 guideline. Cytotherapy. 2018;20(6):806–19.

    Article  PubMed  Google Scholar 

  80. Park SS. Cell therapy applications for retinal vascular diseases: diabetic retinopathy and retinal vein occlusion. Invest Ophthalmol Vis Sci. 2016;57(5):ORSFj1–ORSFj10.

    Article  CAS  PubMed  Google Scholar 

  81. van der Spoel TI, Vrijsen KR, Koudstaal S, Sluijter JP, Nijsen JF, de Jong HW, et al. Transendocardial cell injection is not superior to intracoronary infusion in a porcine model of ischaemic cardiomyopathy: a study on delivery efficiency. J Cell Mol Med. 2012;16(11):2768–76.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Kanelidis AJ, Premer C, Lopez J, Balkan W, Hare JM. Route of delivery modulates the efficacy of mesenchymal stem cell therapy for myocardial infarction: a meta-analysis of preclinical studies and clinical trials. Circ Res. 2017;120(7):1139–50.

    Article  PubMed  CAS  Google Scholar 

  83. Keshtkar S, Azarpira N, Ghahremani MH. Mesenchymal stem cell-derived extracellular vesicles: novel frontiers in regenerative medicine. Stem Cell Res Ther. 2018;9(1):63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Caplan AI, Dennis JE. Mesenchymal stem cells as trophic mediators. J Cell Biochem. 2006;98(5):1076–84.

    Article  CAS  PubMed  Google Scholar 

  85. Caplan AI, Correa D. The MSC: an injury drugstore. Cell Stem Cell. 2011;9(1):11–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Wu Y, Chen L, Scott PG, Tredget EE. Mesenchymal stem cells enhance wound healing through differentiation and angiogenesis. Stem Cells. 2007;25(10):2648–59.

    Article  CAS  PubMed  Google Scholar 

  87. Bronckaers A, Hilkens P, Martens W, Gervois P, Ratajczak J, Struys T, et al. Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate angiogenesis. Pharmacol Ther. 2014;143(2):181–96.

    Article  CAS  PubMed  Google Scholar 

  88. Kean TJ, Lin P, Caplan AI, Dennis JE. MSCs: delivery routes and engraftment, cell-targeting strategies, and immune modulation. Stem Cells Int. 2013;2013:732742.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Chang CL, Leu S, Sung HC, Zhen YY, Cho CL, Chen A, et al. Impact of apoptotic adipose-derived mesenchymal stem cells on attenuating organ damage and reducing mortality in rat sepsis syndrome induced by cecal puncture and ligation. J Transl Med. 2012;10:244.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Goncalves FDC, Luk F, Korevaar SS, Bouzid R, Paz AH, Lopez-Iglesias C, et al. Membrane particles generated from mesenchymal stromal cells modulate immune responses by selective targeting of pro-inflammatory monocytes. Sci Rep. 2017;7(1):12100.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Luk F, de Witte SF, Korevaar SS, Roemeling-van Rhijn M, Franquesa M, Strini T, et al. Inactivated mesenchymal stem cells maintain immunomodulatory capacity. Stem Cells Dev. 2016;25(18):1342–54.

    Article  CAS  PubMed  Google Scholar 

  92. Howard D, Buttery LD, Shakesheff KM, Roberts SJ. Tissue engineering: strategies, stem cells and scaffolds. J Anat. 2008;213(1):66–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Petrella F, Spaggiari L, Acocella F, Barberis M, Bellomi M, Brizzola S, et al. Airway fistula closure after stem-cell infusion. N Engl J Med. 2015;372(1):96–7.

    Article  PubMed  Google Scholar 

  94. Luk F, Carreras-Planella L, Korevaar SS, de Witte SFH, Borras FE, Betjes MGH, et al. Inflammatory conditions dictate the effect of mesenchymal stem or stromal cells on B cell function. Front Immunol. 2017;8:1042.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  95. Wu Y, Hoogduijn MJ, Baan CC, Korevaar SS, de Kuiper R, Yan L, et al. Adipose tissue-derived mesenchymal stem cells have a heterogenic cytokine secretion profile. Stem Cells Int. 2017;2017:4960831.

    Article  PubMed  PubMed Central  Google Scholar 

  96. De Witte SFH, Peters FS, Merino A, Korevaar SS, Van Meurs JBJ, O'Flynn L, et al. Epigenetic changes in umbilical cord mesenchymal stromal cells upon stimulation and culture expansion. Cytotherapy. 2018;20(7):919–29.

    Article  PubMed  CAS  Google Scholar 

  97. Melief SM, Geutskens SB, Fibbe WE, Roelofs H. Multipotent stromal cells skew monocytes towards an anti-inflammatory interleukin-10-producing phenotype by production of interleukin-6. Haematologica. 2013;98(6):888–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Nemeth K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, Doi K, et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med. 2009;15(1):42–9.

    Article  CAS  PubMed  Google Scholar 

  99. Ko JH, Lee HJ, Jeong HJ, Kim MK, Wee WR, Yoon SO, et al. Mesenchymal stem/stromal cells precondition lung monocytes/macrophages to produce tolerance against Allo- and autoimmunity in the eye. Proc Natl Acad Sci U S A. 2016;113(1):158–63.

    Article  CAS  PubMed  Google Scholar 

  100. Krasnodembskaya A, Samarani G, Song Y, Zhuo H, Su X, Lee JW, et al. Human mesenchymal stem cells reduce mortality and bacteremia in gram-negative sepsis in mice in part by enhancing the phagocytic activity of blood monocytes. Am J Physiol Lung Cell Mol Physiol. 2012;302(10):L1003–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Braza F, Dirou S, Forest V, Sauzeau V, Hassoun D, Chesne J, et al. Mesenchymal stem cells induce suppressive macrophages through phagocytosis in a mouse model of asthma. Stem Cells. 2016;34(7):1836–45.

    Article  CAS  PubMed  Google Scholar 

  102. Ge W, Jiang J, Arp J, Liu W, Garcia B, Wang H. Regulatory T-cell generation and kidney allograft tolerance induced by mesenchymal stem cells associated with indoleamine 2,3-dioxygenase expression. Transplantation. 2010;90(12):1312–20.

    Article  CAS  PubMed  Google Scholar 

  103. Miteva K, Pappritz K, El-Shafeey M, Dong F, Ringe J, Tschope C, et al. Mesenchymal stromal cells modulate monocytes trafficking in Coxsackievirus B3-induced myocarditis. Stem Cells Transl Med. 2017;6(4):1249–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Duffy MM, Ritter T, Ceredig R, Griffin MD. Mesenchymal stem cell effects on T-cell effector pathways. Stem Cell Res Ther. 2011;2(4):34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995;155(3):1151–64.

    Article  CAS  PubMed  Google Scholar 

  106. Khosravi M, Karimi MH, Hossein Aghdaie M, Kalani M, Naserian S, Bidmeshkipour A. Mesenchymal stem cells can induce regulatory T cells via modulating miR-126a but not miR-10a. Gene. 2017;627:327–36.

    Article  CAS  PubMed  Google Scholar 

  107. Gieseke F, Bohringer J, Bussolari R, Dominici M, Handgretinger R, Muller I. Human multipotent mesenchymal stromal cells use galectin-1 to inhibit immune effector cells. Blood. 2010;116(19):3770–9.

    Article  CAS  PubMed  Google Scholar 

  108. Rosser EC, Mauri C. Regulatory B cells: origin, phenotype, and function. Immunity. 2015;42(4):607–12.

    Article  CAS  PubMed  Google Scholar 

  109. Franquesa M, Mensah FK, Huizinga R, Strini T, Boon L, Lombardo E, et al. Human adipose tissue-derived mesenchymal stem cells abrogate plasmablast formation and induce regulatory B cells independently of T helper cells. Stem Cells. 2015;33(3):880–91.

    Article  CAS  PubMed  Google Scholar 

  110. Luz-Crawford P, Djouad F, Toupet K, Bony C, Franquesa M, Hoogduijn MJ, et al. Mesenchymal stem cell-derived interleukin 1 receptor antagonist promotes macrophage polarization and inhibits B cell differentiation. Stem Cells. 2016;34(2):483–92.

    Article  CAS  PubMed  Google Scholar 

  111. Deng Y, Zhang Y, Ye L, Zhang T, Cheng J, Chen G, et al. Umbilical cord-derived mesenchymal stem cells instruct monocytes towards an IL10-producing phenotype by secreting IL6 and HGF. Sci Rep. 2016;6:37566.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Sato K, Ozaki K, Oh I, Meguro A, Hatanaka K, Nagai T, et al. Nitric oxide plays a critical role in suppression of T-cell proliferation by mesenchymal stem cells. Blood. 2007;109(1):228–34.

    Article  CAS  PubMed  Google Scholar 

  113. Spaggiari GM, Capobianco A, Abdelrazik H, Becchetti F, Mingari MC, Moretta L. Mesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity, and cytokine production: role of indoleamine 2,3-dioxygenase and prostaglandin E2. Blood. 2008;111(3):1327–33.

    Article  CAS  PubMed  Google Scholar 

  114. Moll G, Rasmusson-Duprez I, von Bahr L, Connolly-Andersen AM, Elgue G, Funke L, et al. Are therapeutic human mesenchymal stromal cells compatible with human blood? Stem Cells. 2012;30(7):1565–74.

    Article  CAS  PubMed  Google Scholar 

  115. Moll G, Ignatowicz L, Catar R, Luecht C, Sadeghi B, Hamad O, et al. Different Procoagulant activity of therapeutic mesenchymal stromal cells derived from bone marrow and placental decidua. Stem Cells Dev. 2015;24(19):2269–79.

    Article  CAS  PubMed  Google Scholar 

  116. Christy BA, Herzig MC, Montgomery RK, Delavan C, Bynum JA, Reddoch KM, et al. Procoagulant activity of human mesenchymal stem cells. J Trauma Acute Care Surg. 2017;83(1 Suppl 1):S164–S9.

    Article  CAS  PubMed  Google Scholar 

  117. Liao L, Shi B, Chang H, Su X, Zhang L, Bi C, et al. Heparin improves BMSC cell therapy: anticoagulant treatment by heparin improves the safety and therapeutic effect of bone marrow-derived mesenchymal stem cell cytotherapy. Theranostics. 2017;7(1):106–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Moll G, Ankrum JA, Kamhieh-Milz J, Bieback K, Ringden O, Volk HD, et al. Intravascular mesenchymal stromal/stem cell therapy product diversification: time for new clinical guidelines. Trends Mol Med. 2019;25(2):149–63.

    Article  PubMed  Google Scholar 

  119. Roobrouck VD, Clavel C, Jacobs SA, Ulloa-Montoya F, Crippa S, Sohni A, et al. 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(5):871–82.

    Article  CAS  PubMed  Google Scholar 

  120. Gruber R, Kandler B, Holzmann P, Vogele-Kadletz M, Losert U, Fischer MB, et al. Bone marrow stromal cells can provide a local environment that favors migration and formation of tubular structures of endothelial cells. Tissue Eng. 2005;11(5–6):896–903.

    Article  CAS  PubMed  Google Scholar 

  121. Kinnaird T, Stabile E, Burnett MS, Lee CW, Barr S, Fuchs S, et al. Marrow-derived stromal cells express genes encoding a broad spectrum of arteriogenic cytokines and promote in vitro and in vivo arteriogenesis through paracrine mechanisms. Circ Res. 2004;94(5):678–85.

    Article  CAS  PubMed  Google Scholar 

  122. Crisostomo PR, Wang Y, Markel TA, Wang M, Lahm T, Meldrum DR. Human mesenchymal stem cells stimulated by TNF-alpha, LPS, or hypoxia produce growth factors by an NF kappa B- but not JNK-dependent mechanism. Am J Physiol Cell Physiol. 2008;294(3):C675–82.

    Article  CAS  PubMed  Google Scholar 

  123. Whyte JL, Ball SG, Shuttleworth CA, Brennan K, Kielty CM. Density of human bone marrow stromal cells regulates commitment to vascular lineages. Stem Cell Res. 2011;6(3):238–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Takahashi M, Suzuki E, Oba S, Nishimatsu H, Kimura K, Nagano T, et al. Adipose tissue-derived stem cells inhibit neointimal formation in a paracrine fashion in rat femoral artery. Am J Physiol Heart Circ Physiol. 2010;298(2):H415–23.

    Article  CAS  PubMed  Google Scholar 

  125. Thum T, Bauersachs J, Poole-Wilson PA, Volk HD, Anker SD. The dying stem cell hypothesis: immune modulation as a novel mechanism for progenitor cell therapy in cardiac muscle. J Am Coll Cardiol. 2005;46(10):1799–802.

    Article  CAS  PubMed  Google Scholar 

  126. Weiss ARR, Dahlke MH. Immunomodulation by Mesenchymal Stem Cells (MSCs): mechanisms of action of living, apoptotic, and dead MSCs. Front Immunol. 2019;10:1191.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Sung PH, Chang CL, Tsai TH, Chang LT, Leu S, Chen YL, et al. Apoptotic adipose-derived mesenchymal stem cell therapy protects against lung and kidney injury in sepsis syndrome caused by cecal ligation puncture in rats. Stem Cell Res Ther. 2013;4(6):155.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Bruno S, Grange C, Deregibus MC, Calogero RA, Saviozzi S, Collino F, et al. Mesenchymal stem cell-derived microvesicles protect against acute tubular injury. J Am Soc Nephrol. 2009;20(5):1053–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Li T, Yan Y, Wang B, Qian H, Zhang X, Shen L, et al. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate liver fibrosis. Stem Cells Dev. 2013;22(6):845–54.

    Article  CAS  PubMed  Google Scholar 

  130. Arslan F, Lai RC, Smeets MB, Akeroyd L, Choo A, Aguor EN, et al. Mesenchymal stem cell-derived exosomes increase ATP levels, decrease oxidative stress and activate PI3K/Akt pathway to enhance myocardial viability and prevent adverse remodeling after myocardial ischemia/reperfusion injury. Stem Cell Res. 2013;10(3):301–12.

    Article  CAS  PubMed  Google Scholar 

  131. Xin H, Li Y, Cui Y, Yang JJ, Zhang ZG, Chopp M. Systemic administration of exosomes released from mesenchymal stromal cells promote functional recovery and neurovascular plasticity after stroke in rats. J Cereb Blood Flow Metab. 2013;33(11):1711–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Rani S, Ryan AE, Griffin MD, Ritter T. Mesenchymal stem cell-derived extracellular vesicles: toward cell-free therapeutic applications. Mol Ther. 2015;23(5):812–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Wu HH, Zhou Y, Tabata Y, Gao JQ. Mesenchymal stem cell-based drug delivery strategy: from cells to biomimetic. J Control Release. 2019;294:102–13.

    Article  CAS  PubMed  Google Scholar 

  134. Coccè V, Farronato D, Brini AT, Masia C, Giannì AB, Piovani G, et al. Drug loaded gingival mesenchymal stromal cells (GinPa-MSCs) inhibit in vitro proliferation of Oral squamous cell carcinoma. Sci Rep. 2017;7(1):9376.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Bonomi A, Silini A, Vertua E, Signoroni PB, Coccè V, Cavicchini L, et al. Human amniotic mesenchymal stromal cells (hAMSCs) as potential vehicles for drug delivery in cancer therapy: an in vitro study. Stem Cell Res Ther. 2015;6(1):155.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  136. Pessina A, Coccè V, Pascucci L, Bonomi A, Cavicchini L, Sisto F, et al. Mesenchymal stromal cells primed with paclitaxel attract and kill leukaemia cells, inhibit angiogenesis and improve survival of leukaemia-bearing mice. Br J Haematol. 2013;160(6):766–78.

    Article  CAS  PubMed  Google Scholar 

  137. Cao B, Yang M, Zhu Y, Qu X, Mao C. Stem cells loaded with nanoparticles as a drug carrier for in vivo breast cancer therapy. Adv Mater. 2014;26(27):4627–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Huang X, Neretina S, El-Sayed MA. Gold Nanorods: from synthesis and properties to biological and biomedical applications. Adv Mater. 2009;21(48):4880–910.

    Article  CAS  PubMed  Google Scholar 

  139. Cheng L, Wang C, Ma X, Wang Q, Cheng Y, Wang H, et al. Multifunctional upconversion nanoparticles for dual-modal imaging-guided stem cell therapy under remote magnetic control. Adv Funct Mater. 2013;23(3):272–80.

    Article  CAS  Google Scholar 

  140. Boltze J, Arnold A, Walczak P, Jolkkonen J, Cui L, Wagner DC. The dark side of the force – constraints and complications of cell therapies for stroke. Front Neurol. 2015;6:155.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Duijvestein M, Vos AC, Roelofs H, Wildenberg ME, Wendrich BB, Verspaget HW, et al. Autologous bone marrow-derived mesenchymal stromal cell treatment for refractory luminal Crohn’s disease: results of a phase I study. Gut. 2010;59(12):1662–9.

    Article  PubMed  Google Scholar 

  142. Savolainen H. Encephalopathy, stroke, and myocardial infarction with DMSO use in stem cell transplantation. Neurology. 2007;69(5):494; author reply -5

    Article  PubMed  Google Scholar 

  143. Stolzing A, Jones E, McGonagle D, Scutt A. Age-related changes in human bone marrow-derived mesenchymal stem cells: consequences for cell therapies. Mech Ageing Dev. 2008;129(3):163–73.

    Article  CAS  PubMed  Google Scholar 

  144. Turinetto V, Vitale E, Giachino C. Senescence in human mesenchymal stem cells: functional changes and implications in stem cell-based therapy. Int J Mol Sci. 2016;17(7):6–8.

    Google Scholar 

  145. Bonab MM, Alimoghaddam K, Talebian F, Ghaffari SH, Ghavamzadeh A, Nikbin B. Aging of mesenchymal stem cell in vitro. BMC Cell Biol. 2006;7:14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  146. von Bahr L, Sundberg B, Lonnies L, Sander B, Karbach H, Hagglund H, et al. Long-term complications, immunologic effects, and role of passage for outcome in mesenchymal stromal cell therapy. Biol Blood Marrow Transplant. 2012;18(4):557–64.

    Article  Google Scholar 

  147. Cornelio DA, Tavares JC, Pimentel TV, Cavalcanti GB Jr, Batistuzzo de Medeiros SR. Cytokinesis-block micronucleus assay adapted for analyzing genomic instability of human mesenchymal stem cells. Stem Cells Dev. 2014;23(8):823–38.

    Article  CAS  PubMed  Google Scholar 

  148. Wagner J, Kean T, Young R, Dennis JE, Caplan AI. Optimizing mesenchymal stem cell-based therapeutics. Curr Opin Biotechnol. 2009;20(5):531–6.

    Article  CAS  PubMed  Google Scholar 

  149. Jung JW, Kwon M, Choi JC, Shin JW, Park IW, Choi BW, et al. Familial occurrence of pulmonary embolism after intravenous, adipose tissue-derived stem cell therapy. Yonsei Med J. 2013;54(5):1293–6.

    Article  PubMed  PubMed Central  Google Scholar 

  150. Wu Z, Zhang S, Zhou L, Cai J, Tan J, Gao X, et al. Thromboembolism induced by umbilical cord mesenchymal stem cell infusion: a report of two cases and literature review. Transplant Proc. 2017;49(7):1656–8.

    Article  CAS  PubMed  Google Scholar 

  151. Sokal EM, Stéphenne X, Ottolenghi C, Jazouli N, Clapuyt P, Lacaille F, et al. Liver engraftment and repopulation by in vitro expanded adult derived human liver stem cells in a child with ornithine carbamoyltransferase deficiency. JIMD Rep. 2014;13:65–72.

    Article  PubMed  Google Scholar 

  152. Toma C, Wagner WR, Bowry S, Schwartz A, Villanueva F. Fate of culture-expanded mesenchymal stem cells in the microvasculature: in vivo observations of cell kinetics. Circ Res. 2009;104(3):398–402.

    Article  CAS  PubMed  Google Scholar 

  153. Gleeson BM, Martin K, Ali MT, Kumar AHS, Pillai MG-K, Kumar SPG, et al. Bone marrow-derived mesenchymal stem cells have innate Procoagulant activity and cause microvascular obstruction following intracoronary delivery: amelioration by Antithrombin therapy. Stem Cells. 2015;33(9):2726–37.

    Article  CAS  PubMed  Google Scholar 

  154. Acosta L, Hmadcha A, Escacena N, Perez-Camacho I, de la Cuesta A, Ruiz-Salmeron R, et al. Adipose mesenchymal stromal cells isolated from type 2 diabetic patients display reduced fibrinolytic activity. Diabetes. 2013;62(12):4266–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Coppin L, Sokal E, Stéphenne X. Thrombogenic risk induced by intravascular mesenchymal stem cell therapy: current status and future perspectives. Cell. 2019;8(10):1160.

    Article  CAS  Google Scholar 

  156. Melmed GY, Pandak WM, Casey K, Abraham B, Valentine J, Schwartz D, et al. Human placenta-derived cells (PDA-001) for the treatment of moderate-to-severe Crohn’s disease: a phase 1b/2a study. Inflamm Bowel Dis. 2015;21(8):1809–16.

    Article  PubMed  Google Scholar 

  157. Baygan A, Aronsson-Kurttila W, Moretti G, Tibert B, Dahllöf G, Klingspor L, et al. Safety and side effects of using placenta-derived Decidual stromal cells for graft-versus-host disease and hemorrhagic cystitis. Front Immunol. 2017;8:795.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  158. Kaipe H, Carlson LM, Erkers T, Nava S, Mollden P, Gustafsson B, et al. Immunogenicity of decidual stromal cells in an epidermolysis bullosa patient and in allogeneic hematopoietic stem cell transplantation patients. Stem Cells Dev. 2015;24(12):1471–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Lohan P, Treacy O, Griffin MD, Ritter T, Ryan AE. Anti-donor immune responses elicited by allogeneic mesenchymal stem cells and their extracellular vesicles: are we still learning? Front Immunol. 2017;8:1626.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  160. Steward O, Sharp KG, Yee KM, Hatch MN, Bonner JF. Characterization of ectopic colonies that form in widespread areas of the nervous system with neural stem cell transplants into the site of a severe spinal cord injury. J Neurosci. 2014;34(42):14013–21.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  161. Kawai H, Yamashita T, Ohta Y, Deguchi K, Nagotani S, Zhang X, et al. Tridermal tumorigenesis of induced pluripotent stem cells transplanted in ischemic brain. J Cereb Blood Flow Metab. 2010;30(8):1487–93.

    Article  PubMed  PubMed Central  Google Scholar 

  162. Erdo F, Buhrle C, Blunk J, Hoehn M, Xia Y, Fleischmann B, et al. Host-dependent tumorigenesis of embryonic stem cell transplantation in experimental stroke. J Cereb Blood Flow Metab. 2003;23(7):780–5.

    Article  PubMed  Google Scholar 

  163. Minieri V, Saviozzi S, Gambarotta G, Lo Iacono M, Accomasso L, Cibrario Rocchietti E, et al. Persistent DNA damage-induced premature senescence alters the functional features of human bone marrow mesenchymal stem cells. J Cell Mol Med. 2015;19(4):734–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Schu S, Nosov M, O'Flynn L, Shaw G, Treacy O, Barry F, et al. Immunogenicity of allogeneic mesenchymal stem cells. J Cell Mol Med. 2012;16(9):2094–103.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Posel C, Scheibe J, Kranz A, Bothe V, Quente E, Frohlich W, et al. Bone marrow cell transplantation time-dependently abolishes efficacy of granulocyte colony-stimulating factor after stroke in hypertensive rats. Stroke. 2014;45(8):2431–7.

    Article  PubMed  CAS  Google Scholar 

  166. Chen X, Gan Y, Li W, Su J, Zhang Y, Huang Y, et al. The interaction between mesenchymal stem cells and steroids during inflammation. Cell Death Dis. 2014;5:e1009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Liu C, Fan Y, Zhou L, Zhu HY, Song YC, Hu L, et al. Pretreatment of mesenchymal stem cells with angiotensin II enhances paracrine effects, angiogenesis, gap junction formation and therapeutic efficacy for myocardial infarction. Int J Cardiol. 2015;188:22–32.

    Article  PubMed  Google Scholar 

  168. Liu J, Zhu P, Song P, Xiong W, Chen H, Peng W, et al. Pretreatment of adipose derived stem cells with curcumin facilitates myocardial recovery via Antiapoptosis and angiogenesis. Stem Cells Int. 2015;2015:638153.

    Article  PubMed  PubMed Central  Google Scholar 

  169. Wei ZZ, Zhu YB, Zhang JY, McCrary MR, Wang S, Zhang YB, et al. Priming of the cells: hypoxic preconditioning for stem cell therapy. Chin Med J. 2017;130(19):2361–74.

    PubMed  PubMed Central  CAS  Google Scholar 

  170. Liu ZJ, Daftarian P, Kovalski L, Wang B, Tian R, Castilla DM, et al. Directing and potentiating stem cell-mediated angiogenesis and tissue repair by cell surface E-selectin coating. PLoS One. 2016;11(4):e0154053.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2021 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Lopes, L.L.N.M., Navarro, T.P., Dardik, A. (2021). Introduction to Stem Cell Therapy and Its Application in Vascular Diseases. In: Navarro, T.P., Minchillo Lopes, L.L.N., Dardik, A. (eds) Stem Cell Therapy for Vascular Diseases. Springer, Cham. https://doi.org/10.1007/978-3-030-56954-9_1

Download citation

  • DOI: https://doi.org/10.1007/978-3-030-56954-9_1

  • Published:

  • Publisher Name: Springer, Cham

  • Print ISBN: 978-3-030-56953-2

  • Online ISBN: 978-3-030-56954-9

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics