Skip to main content
Log in

Mesenchymal Stem Cell Transplantation for Ischemic Diseases: Mechanisms and Challenges

  • Review Article
  • Published:
Tissue Engineering and Regenerative Medicine Aims and scope

Abstract

Ischemic diseases are conditions associated with the restriction or blockage of blood supply to specific tissues. These conditions can cause moderate to severe complications in patients, and can lead to permanent disabilities. Since they are blood vessel-related diseases, ischemic diseases are usually treated with endothelial cells or endothelial progenitor cells that can regenerate new blood vessels. However, in recent years, mesenchymal stem cells (MSCs) have shown potent bioeffects on angiogenesis, thus playing a role in blood regeneration. Indeed, MSCs can trigger angiogenesis at ischemic sites by several mechanisms related to their trans-differentiation potential. These mechanisms include inhibition of apoptosis, stimulation of angiogenesis via angiogenic growth factors, and regulation of immune responses, as well as regulation of scarring to suppress blood vessel regeneration when needed. However, preclinical and clinical trials of MSC transplantation in ischemic diseases have shown some limitations in terms of treatment efficacy. Such studies have emphasized the current challenges of MSC-based therapies. Treatment efficacy could be enhanced if the limitations were better understood and potentially resolved. This review will summarize some of the strategies by which MSCs have been utilized for ischemic disease treatment, and will highlight some challenges of those applications as well as suggesting some strategies to improve treatment efficacy.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

modified by the presence of MSC administration (blue arrow). Changes in the environment such as oxygen and nutrition deprivation and host immune response can negatively affect the survival and proliferation of MSCs. On the other hand, the administration of MSCs during ischemic condition can help to reverse the damages caused by ischemic condition. Improvement of the condition is induced by the effect generated from MSCs characteristics of immunomodulation, microenvironment generation, homing and differentiation. MSCs generate secretome containing growth factors, cytokines and chemokines to modulate the host immune response, resolving cell death signal and balancing angiogenic and angiostatic pool in the ischemic site. MSCs can also home and guide the regeneration of nerve in the ischemic tissue to support tissue recovery

Similar content being viewed by others

References

  1. Calvert JW. Chapter 5 - Ischemic heart disease and its consequences. In: Willis MS, Homeister JW, Stone JR, editors. Cellular and molecular pathobiology of cardiovascular disease. San Diego: Academic Press; 2014. p. 79–100.

    Chapter  Google Scholar 

  2. Fowkes FG, Rudan D, Rudan I, Aboyans V, Denenberg JO, McDermott MM, et al. Comparison of global estimates of prevalence and risk factors for peripheral artery disease in 2000 and 2010: a systematic review and analysis. Lancet. 2013;382:1329–40.

    Article  PubMed  Google Scholar 

  3. Li S, Wang X, Li J, Zhang J, Zhang F, Hu J, et al. Advances in the treatment of ischemic diseases by mesenchymal stem cells. Stem Cells Int. 2016;2016:5896061.

    PubMed  PubMed Central  Google Scholar 

  4. Jeong H, Yim HW, Park HJ, Cho Y, Hong H, Kim NJ, et al. Mesenchymal stem cell therapy for ischemic heart disease: systematic review and meta-analysis. Int J Stem Cells. 2018;11:1–12.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Wang F, Tang H, Zhu J, Zhang JH. Transplanting mesenchymal stem cells for treatment of ischemic stroke. Cell Transplant. 2018;27:1825–34.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Gupta PK, Chullikana A, Parakh R, Desai S, Das A, Gottipamula S, et al. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lee JS, Hong JM, Moon GJ, Lee PH, Ahn YH, Bang OY. A long-term follow-up study of intravenous autologous mesenchymal stem cell transplantation in patients with ischemic stroke. Stem cells. 2010;28:1099–106.

    Article  PubMed  Google Scholar 

  8. Lalu MM, McIntyre L, Pugliese C, Fergusson D, Winston BW, Marshall JC, et al. Safety of cell therapy with mesenchymal stromal cells (SafeCell): a systematic review and meta-analysis of clinical trials. PLoS One. 2012;7:e47559.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Prasad K, Sharma A, Garg A, Mohanty S, Bhatnagar S, Johri S, et al. Intravenous autologous bone marrow mononuclear stem cell therapy for ischemic stroke: a multicentric, randomized trial. Stroke. 2014;45:3618–24.

    Article  CAS  PubMed  Google Scholar 

  10. Silva LHA, Antunes MA, Dos Santos CC, Weiss DJ, Cruz FF, Rocco PRM. Strategies to improve the therapeutic effects of mesenchymal stromal cells in respiratory diseases. Stem Cell Res Ther. 2018;9:45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Li L, Chen X, Wang WE, Zeng C. How to improve the survival of transplanted mesenchymal stem cell in ischemic heart? Stem Cells Int. 2016;2016:9682757.

    Article  PubMed  Google Scholar 

  12. Luo R, Lu Y, Liu J, Cheng J, Chen Y. Enhancement of the efficacy of mesenchymal stem cells in the treatment of ischemic diseases. Biomed Pharmacother. 2019;109:2022–34.

    Article  CAS  PubMed  Google Scholar 

  13. Kim SH, Cho JH, Lee YH, Lee JH, Kim SS, Kim MY, et al. Improvement in left ventricular function with intracoronary mesenchymal stem cell therapy in a patient with anterior wall ST-segment elevation myocardial infarction. Cardiovasc Drugs Ther. 2018;32:329–38.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Penn MS, Ellis S, Gandhi S, Greenbaum A, Hodes Z, Mendelsohn FO, et al. Adventitial delivery of an allogeneic bone marrow-derived adherent stem cell in acute myocardial infarction: phase I clinical study. Circ Res. 2012;110:304–11.

    Article  CAS  PubMed  Google Scholar 

  15. Mathiasen AB, Qayyum AA, Jørgensen E, Helqvist S, Fischer-Nielsen A, Kofoed KF, et al. Bone marrow-derived mesenchymal stromal cell treatment in patients with severe ischaemic heart failure: a randomized placebo-controlled trial (MSC-HF trial). Eur Heart J. 2015;36:1744–53.

    Article  CAS  PubMed  Google Scholar 

  16. Mathiasen AB, Qayyum AA, Jørgensen E, Helqvist S, Kofoed KF, Haack-Sørensen M, et al. Bone marrow-derived mesenchymal stromal cell treatment in patients with ischaemic heart failure: final 4-year follow-up of the MSC-HF trial. Eur J Heart Fail. 2020;22:884–92.

    Article  CAS  PubMed  Google Scholar 

  17. Ulus AT, Mungan C, Kurtoglu M, Celikkan FT, Akyol M, Sucu M, et al. Intramyocardial transplantation of umbilical cord mesenchymal stromal cells in chronic ischemic cardiomyopathy: a controlled, randomized clinical trial (HUC-HEART trial). Int J Stem Cells. 2020;13:364–76.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Xiao W, Guo S, Gao C, Dai G, Gao Y, Li M, et al. A Randomized comparative study on the efficacy of intracoronary infusion of autologous bone marrow mononuclear cells and mesenchymal stem cells in patients with dilated cardiomyopathy. Int Heart J. 2017;58:238–44.

    Article  CAS  PubMed  Google Scholar 

  19. Qayyum AA, Mathiasen AB, Mygind ND, Vejlstrup NG, Kastrup J. Cardiac magnetic resonance imaging used for evaluation of adipose-derived stromal cell therapy in patients with chronic ischemic heart disease. Cell Transplant. 2019;28:1700–8.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Florea V, Rieger AC, DiFede DL, El-Khorazaty J, Natsumeda M, Banerjee MN, et al. Dose comparison study of allogeneic mesenchymal stem cells in patients with ischemic cardiomyopathy (The TRIDENT Study). Circ Res. 2017;121:1279–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Hare JM, Fishman JE, Gerstenblith G, DiFede Velazquez DL, Zambrano JP, Suncion VY, et al. Comparison of allogeneic vs autologous bone marrow–derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. JAMA. 2012;308:2369–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Gralla J, Brekenfeld C, Mordasini P, Schroth G. Mechanical thrombolysis and stenting in acute ischemic stroke. Stroke. 2012;43:280–5.

    Article  PubMed  Google Scholar 

  23. Khazaei M, Davoodian A, Taheri M, Ghafouri-Fard S. Former antiplatelet drug administration and consequences of intravenous thrombolysis in acute ischemic stroke. Hum Antibodies. 2020;28:53–6.

    Article  PubMed  Google Scholar 

  24. Su YH, Chen CH, Lin HJ, Chen YW, Tseng MC, Hsieh HC, et al. Safety and effectiveness of intravenous thrombolysis for acute ischemic stroke outside the coverage of national health insurance in Taiwan. Acta Neurol Taiwan. 2017;26:3–12.

    PubMed  Google Scholar 

  25. Honmou O, Houkin K, Matsunaga T, Niitsu Y, Ishiai S, Onodera R, et al. Intravenous administration of auto serum-expanded autologous mesenchymal stem cells in stroke. Brain: J Neurol. 2011;134:1790–807.

    Article  Google Scholar 

  26. Steinberg GK, Kondziolka D, Wechsler LR, Lunsford LD, Kim AS, Johnson JN, et al. Two-year safety and clinical outcomes in chronic ischemic stroke patients after implantation of modified bone marrow-derived mesenchymal stem cells (SB623): a phase 1/2a study. J Neurosurg. 2018;23:1–11.

    Google Scholar 

  27. Levy ML, Crawford JR, Dib N, Verkh L, Tankovich N, Cramer SC. Phase I/II study of safety and preliminary efficacy of intravenous allogeneic mesenchymal stem cells in chronic stroke. Stroke. 2019;50:2835–41.

    Article  PubMed  Google Scholar 

  28. Jaillard A, Hommel M, Moisan A, Zeffiro TA, Favre-Wiki IM, Barbieux-Guillot M, et al. Autologous mesenchymal stem cells improve motor recovery in subacute ischemic stroke: a randomized clinical trial. Transl Stroke Res. 2020;11:910–23.

    Article  CAS  PubMed  Google Scholar 

  29. Lu D, Jiang Y, Deng W, Zhang Y, Liang Z, Wu Q, et al. Long-Term outcomes of BMMSC compared with BMMNC for treatment of critical limb ischemia and foot ulcer in patients with diabetes. Cell Transplant. 2019;28:645–52.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Gupta PK, Krishna M, Chullikana A, Desai S, Murugesan R, Dutta S, et al. Administration of adult human bone marrow-derived, cultured, pooled, allogeneic mesenchymal stromal cells in critical limb ischemia due to Buerger’s disease: phase II study report suggests clinical efficacy. Stem Cells Transl Med. 2017;6:689–99.

    Article  PubMed  Google Scholar 

  31. Gao WH, Gao HY, Li YT, Huang PP. Effectiveness of umbilical cord mesenchymal stem cells in patients with critical limb ischemia. Med Clin. 2019;153:341–6.

    Article  Google Scholar 

  32. Huang P. Therapeutic outcomes of transplanting human umbilical cord mesenchymal stem cells in patients with critical limb ischemia. Eur J Vasc Endovasc Surg. 2019;58:e388–9.

    Article  Google Scholar 

  33. Wang J, Zeng XX, Cai W, Han ZB, Zhu LY, Liu JY, et al. Safety and efficacy of placenta-derived mesenchymal stem cell treatment for diabetic patients with critical limb ischemia: a pilot study. Experim Clin Endocrinol Diabetes. 2019;1:1.

    Google Scholar 

  34. Heldman AW, DiFede DL, Fishman JE, Zambrano JP, Trachtenberg BH, Karantalis V, et al. Transendocardial mesenchymal stem cells and mononuclear bone marrow cells for ischemic cardiomyopathy: the TAC-HFT randomized trial. JAMA. 2014;311:62–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lee HC, An SG, Lee HW, Park JS, Cha KS, Hong TJ, et al. Safety and effect of adipose tissue-derived stem cell implantation in patients with critical limb ischemia: a pilot study. Circ J. 2012;76:1750–60.

    Article  CAS  PubMed  Google Scholar 

  36. Salter B, Sehmi R. The role of bone marrow-derived endothelial progenitor cells and angiogenic responses in chronic obstructive pulmonary disease. J Thor Dis. 2017;9:2168–77.

    Article  Google Scholar 

  37. Bakondi B, Shimada IS, Perry A, Munoz JR, Ylostalo J, Howard AB, et al. CD133 identifies a human bone marrow stem/progenitor cell sub-population with a repertoire of secreted factors that protect against stroke. Mol Ther. 2009;17:1938–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Nasseri BA, Ebell W, Dandel M, Kukucka M, Gebker R, Doltra A, et al. Autologous CD133+ bone marrow cells and bypass grafting for regeneration of ischaemic myocardium: the Cardio133 trial. Eur Heart J. 2014;35:1263–74.

    Article  CAS  PubMed  Google Scholar 

  39. Forcillo J, Stevens LM, Mansour S, Prieto I, Salem R, Baron C, et al. Implantation of CD133+ stem cells in patients undergoing coronary bypass surgery: IMPACT-CABG pilot trial. Can J Cardiol. 2013;29:441–7.

    Article  PubMed  Google Scholar 

  40. Stamm C, Kleine HD, Choi YH, Dunkelmann S, Lauffs JA, Lorenzen B, et al. Intramyocardial delivery of CD133+ bone marrow cells and coronary artery bypass grafting for chronic ischemic heart disease: safety and efficacy studies. J Thorac Cardiovasc Surg. 2007;133:717–25.

    Article  PubMed  Google Scholar 

  41. Naseri MH, Madani H, Ahmadi Tafti SH, Moshkani Farahani M, Kazemi Saleh D, Hosseinnejad H, et al. COMPARE CPM-RMI trial: intramyocardial transplantation of autologous bone marrow-derived CD133+ cells and MNCs during CABG in patients with recent MI: a phase II/III, multicenter, placebo-controlled, randomized, Double-Blind Clinical Trial . Cell J. 2018;20:267–77.

    PubMed  PubMed Central  Google Scholar 

  42. Makkar RR, Smith RR, Cheng K, Malliaras K, Thomson LE, Berman D, et al. Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (CADUCEUS): a prospective, randomised phase 1 trial. Lancet. 2012;379:895–904.

    Article  Google Scholar 

  43. Chugh AR, Beache GM, Loughran JH, Mewton N, Elmore JB, Kajstura J, et al. Administration of cardiac stem cells in patients with ischemic cardiomyopathy: the SCIPIO trial: surgical aspects and interim analysis of myocardial function and viability by magnetic resonance. Circulation. 2012;126:S54-64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Mou Y, Yue Z, Zhang H, Shi X, Zhang M, Chang X, et al. High quality in vitro expansion of human endothelial progenitor cells of human umbilical vein origin. Int J Med Sci. 2017;14:294–301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. O’Neill DW, Jiang Y, Leary E, Yavanian G, Eminli S, Marasco WA. A Lin-CD45-CD34+ population of extracellular vesicles in human blood that mimics very small embryonic-like stem cells (VSELs) by flow cytometry. Blood. 2012;120:4750.

    Article  Google Scholar 

  46. Sovalat H, Scrofani M, Eidenschenk A, Pasquet S, Rimelen V, Hénon P. Identification and isolation from either adult human bone marrow or G-CSF−mobilized peripheral blood of CD34+/CD133+/CXCR4+/ Lin−CD45− cells, featuring morphological, molecular, and phenotypic characteristics of very small embryonic-like (VSEL) stem cells. Exp Hematol. 2011;39:495–505.

    Article  CAS  PubMed  Google Scholar 

  47. Wang S, Cui J, Peng W, Lu M. Intracoronary autologous CD34+ stem cell therapy for intractable angina. Cardiology. 2010;117:140–7.

    Article  PubMed  Google Scholar 

  48. Velagapudi P, Turagam M, Kolte D, Khera S, Hyder O, Gordon P, et al. Intramyocardial autologous CD34+ cell therapy for refractory angina: A meta-analysis of randomized controlled trials. Cardiovasc Revascular Med: Includ Mol Interv. 2019;20:215–9.

    Article  Google Scholar 

  49. Park M, Seo JJ. Role of HLA in hematopoietic stem cell transplantation. Bone Marrow Res. 2012;2012:680841.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Latham K, Little AM, Madrigal JA. An overview of HLA typing for hematopoietic stem cell transplantation. Methods Mol Biol. 2014;1109:73–85.

    Article  CAS  PubMed  Google Scholar 

  51. Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringdén O. HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol. 2003;31:890–6.

    Article  PubMed  CAS  Google Scholar 

  52. Wang Y, Tian M, Wang F, Heng BC, Zhou J, Cai Z, et al. Understanding the immunological mechanisms of mesenchymal stem cells in allogeneic transplantation: from the aspect of major histocompatibility complex class I. Stem Cells Dev. 2019;28:1141–50.

    Article  PubMed  Google Scholar 

  53. Lee CW, Stabile E, Kinnaird T, Shou M, Devaney JM, Epstein SE, et al. Temporal patterns of gene expression after acute hindlimb ischemia in mice: insights into the genomic program for collateral vessel development. J Am Coll Cardiol. 2004;43:474–82.

    Article  CAS  PubMed  Google Scholar 

  54. Cave Alison C, Ingwall Joanne S, Friedrich J, Liao R, Saupe Kurt W, Apstein Carl S, et al. ATP synthesis during low-flow ischemia. Circulation. 2000;101:2090–6.

    Article  Google Scholar 

  55. Weil MH, Afifi AA. Experimental and clinical studies on lactate and pyruvate as indicators of the severity of acute circulatory failure (shock). Circulation. 1970;41:989–1001.

    Article  CAS  PubMed  Google Scholar 

  56. Chan TS, Cassim S, Raymond VA, Gottschalk S, Merlen G, Zwingmann C, et al. Upregulation of Krebs cycle and anaerobic glycolysis activity early after onset of liver ischemia. PLoS One. 2018;13:e0199177.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Lundberg G, Wahlberg E, Swedenborg J, Sundberg CJ, Ungerstedt U, Olofsson P. Continuous assessment of local metabolism by microdialysis in critical limb ischaemia. Eur J Vascul Endovasc Surg. 2000;19:605–13.

    Article  CAS  Google Scholar 

  58. Bosco G, Yang ZJ, Nandi J, Wang J, Chen C, Camporesi EM. Effects of hyperbaric oxygen on glucose, lactate, glycerol and anti-oxidant enzymes in the skeletal muscle of rats during ischaemia and reperfusion. Clin Exp Pharmacol Physiol. 2007;34:70–6.

    Article  CAS  PubMed  Google Scholar 

  59. Muller M, Schmid R, Nieszpaur-Los M, Fassolt A, Lonnroth P, Fasching P, et al. Key metabolite kinetics in human skeletal muscle during ischaemia and reperfusion: measurement by microdialysis. Eur J Clin Invest. 1995;25:601–7.

    Article  CAS  PubMed  Google Scholar 

  60. Ryan TE, Yamaguchi DJ, Schmidt CA, Zeczycki TN, Shaikh SR, Brophy P, et al. Extensive skeletal muscle cell mitochondriopathy distinguishes critical limb ischemia patients from claudicants. JCI Insight. 2018;3:e123235.

    Article  PubMed Central  Google Scholar 

  61. Sun D, Nguyen N, DeGrado TR, Schwaiger M, Brosius FC 3rd. Ischemia induces translocation of the insulin-responsive glucose transporter GLUT4 to the plasma membrane of cardiac myocytes. Circulation. 1994;89:793–8.

    Article  CAS  PubMed  Google Scholar 

  62. Davey KA, Garlick PB, Warley A, Southworth R. Immunogold labeling study of the distribution of GLUT-1 and GLUT-4 in cardiac tissue following stimulation by insulin or ischemia. Am J Physiol Heart Circ Physiol. 2007;292:H2009–19.

    Article  CAS  PubMed  Google Scholar 

  63. Borsheim E, Tipton KD, Wolf SE, Wolfe RR. Essential amino acids and muscle protein recovery from resistance exercise. Am J Physiol Endocrinol Metab. 2002;283:E648–57.

    Article  CAS  PubMed  Google Scholar 

  64. Drake KJ, Shotwell MS, Wikswo JP, Sidorov VY. Glutamine and glutamate limit the shortening of action potential duration in anoxia-challenged rabbit hearts. Physiol Rep. 2015;3:1.

    Article  CAS  Google Scholar 

  65. Bolotin G, Raman J, Williams U, Bacha E, Kocherginsky M, Jeevanandam V. Glutamine improves myocardial function following ischemia-reperfusion injury. Asian Cardiovasc Thorac Ann. 2007;15:463–7.

    Article  PubMed  Google Scholar 

  66. Liu J, Marchase RB, Chatham JC. Glutamine-induced protection of isolated rat heart from ischemia/reperfusion injury is mediated via the hexosamine biosynthesis pathway and increased protein O-GlcNAc levels. J Mol Cell Cardiol. 2007;42:177–85.

    Article  CAS  PubMed  Google Scholar 

  67. Mohiuddin M, Lee NH, Moon JY, Han WM, Anderson SE, Choi JJ, et al. Critical limb ischemia induces remodeling of skeletal muscle motor unit, myonuclear-, and mitochondrial-domains. Sci Rep. 2019;9:9551.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Tran TP, Tu H, Pipinos II, Muelleman RL, Albadawi H, Li YL. Tourniquet-induced acute ischemia-reperfusion injury in mouse skeletal muscles: Involvement of superoxide. Eur J Pharmacol. 2011;650:328–34.

    Article  CAS  PubMed  Google Scholar 

  69. Ambrosio G, Becker LC, Hutchins GM, Weisman HF, Weisfeldt ML. Reduction in experimental infarct size by recombinant human superoxide dismutase: insights into the pathophysiology of reperfusion injury. Circulation. 1986;74:1424–33.

    Article  CAS  PubMed  Google Scholar 

  70. Kevin LG, Camara AK, Riess ML, Novalija E, Stowe DF. Ischemic preconditioning alters real-time measure of O2 radicals in intact hearts with ischemia and reperfusion. Am J Physiol Heart Circ Physiol. 2003;284:H566–74.

    Article  CAS  PubMed  Google Scholar 

  71. Forbes RA, Steenbergen C, Murphy E. Diazoxide-induced cardioprotection requires signaling through a redox-sensitive mechanism. Circ Res. 2001;88:802–9.

    Article  CAS  PubMed  Google Scholar 

  72. Kirmes I, Szczurek A, Prakash K, Charapitsa I, Heiser C, Musheev M, et al. A transient ischemic environment induces reversible compaction of chromatin. Genome Biol. 2015;16:246.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Batie M, Del Peso L, Rocha S. Hypoxia and chromatin: a focus on transcriptional repression mechanisms. Biomedicines. 2018;6:1.

    Article  CAS  Google Scholar 

  74. Semenza GL. Hypoxia-inducible factor 1: regulator of mitochondrial metabolism and mediator of ischemic preconditioning. Biochem Biophys Acta. 2011;1813:1263–8.

    Article  CAS  PubMed  Google Scholar 

  75. Cai Z, Luo W, Zhan H, Semenza GL. Hypoxia-inducible factor 1 is required for remote ischemic preconditioning of the heart. Proc Natl Acad Sci U S A. 2013;110:17462–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Kapitsinou PP, Sano H, Michael M, Kobayashi H, Davidoff O, Bian A, et al. Endothelial HIF-2 mediates protection and recovery from ischemic kidney injury. J Clin Invest. 2014;124:2396–409.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Sarkar K, Fox-Talbot K, Steenbergen C, Bosch-Marce M, Semenza GL. Adenoviral transfer of HIF-1alpha enhances vascular responses to critical limb ischemia in diabetic mice. Proc Natl Acad Sci U S A. 2009;106:18769–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Pavo N, Lukovic D, Zlabinger K, Zimba A, Lorant D, Goliasch G, et al. Sequential activation of different pathway networks in ischemia-affected and non-affected myocardium, inducing intrinsic remote conditioning to prevent left ventricular remodeling. Sci Rep. 2017;7:43958.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Ganta VC, Choi M, Kutateladze A, Annex BH. VEGF165b modulates endothelial VEGFR1-STAT3 signaling pathway and angiogenesis in human and experimental peripheral arterial disease. Circ Res. 2017;120:282–95.

    Article  CAS  PubMed  Google Scholar 

  80. Zhu H, Xiao F, Wang G, Wei X, Jiang L, Chen Y, et al. STAT3 Regulates self-renewal of adult muscle satellite cells during injury-induced muscle regeneration. Cell Rep. 2016;16:2102–15.

    Article  CAS  PubMed  Google Scholar 

  81. Choi EK, Yeo JS, Park CY, Na H, Lim J, Lee JE, et al. Inhibition of reactive oxygen species downregulates the MAPK pathway in rat spinal cord after limb ischemia reperfusion injury. Int J Surg. 2015;22:74–8.

    Article  PubMed  Google Scholar 

  82. Yang J. The role of reactive oxygen species in angiogenesis and preventing tissue injury after brain ischemia. Microvasc Res. 2019;123:62–7.

    Article  CAS  PubMed  Google Scholar 

  83. Bentley K, Gerhardt H, Bates PA. Agent-based simulation of notch-mediated tip cell selection in angiogenic sprout initialisation. J Theor Biol. 2008;250:25–36.

    Article  CAS  PubMed  Google Scholar 

  84. Moya IM, Umans L, Maas E, Pereira PN, Beets K, Francis A, et al. Stalk cell phenotype depends on integration of Notch and Smad1/5 signaling cascades. Dev Cell. 2012;22:501–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Tetzlaff F, Fischer A. Control of blood vessel formation by notch signaling. Adv Exp Med Biol. 2018;1066:319–38.

    Article  CAS  PubMed  Google Scholar 

  86. Blanco R, Gerhardt H. VEGF and Notch in tip and stalk cell selection. Cold Spring Harbor Perspect Med. 2013;3:a006569.

    Article  CAS  Google Scholar 

  87. Yang Q, Yan W, Li X, Hou L, Dong H, Wang Q, et al. Activation of canonical notch signaling pathway is involved in the ischemic tolerance induced by sevoflurane preconditioning in mice. Anesthesiology. 2012;117:996–1005.

    Article  CAS  PubMed  Google Scholar 

  88. Jin Z, Guo P, Li X, Ke J, Wang Y, Wu H. Neuroprotective effects of irisin against cerebral ischemia/ reperfusion injury via Notch signaling pathway. Biomed Pharmacother. 2019;120:109452.

    Article  CAS  PubMed  Google Scholar 

  89. Dimitrova E, Caromile LA, Laubenbacher R, Shapiro LH. The innate immune response to ischemic injury: a multiscale modeling perspective. BMC Syst Biol. 2018;12:50.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  90. Santos-Zas I, Lemarie J, Tedgui A, Ait-Oufella H. Adaptive immune responses contribute to post-ischemic cardiac remodeling. Front Cardiovasc Med. 2018;5:198.

    Article  CAS  PubMed  Google Scholar 

  91. Rusinkevich V, Huang Y, Chen ZY, Qiang W, Wang YG, Shi YF, et al. Temporal dynamics of immune response following prolonged myocardial ischemia/reperfusion with and without cyclosporine A. Acta Pharmacol Sin. 2019;40:1168–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Yan X, Anzai A, Katsumata Y, Matsuhashi T, Ito K, Endo J, et al. Temporal dynamics of cardiac immune cell accumulation following acute myocardial infarction. J Mol Cell Cardiol. 2013;62:24–35.

    Article  CAS  PubMed  Google Scholar 

  93. Madaro L, Torcinaro A, De Bardi M, Contino FF, Pelizzola M, Diaferia GR, et al. Macrophages fine tune satellite cell fate in dystrophic skeletal muscle of mdx mice. PLoS Genet. 2019;15:e1008408.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Bless NM, Warner RL, Padgaonkar VA, Lentsch AB, Czermak BJ, Schmal H, et al. Roles for C-X-C chemokines and C5a in lung injury after hindlimb ischemia-reperfusion. Am J Physiol. 1999;276:L57-63.

    CAS  PubMed  Google Scholar 

  95. Jaworska K, Ratajczak J, Huang L, Whalen K, Yang M, Stevens BK, et al. Both PD-1 ligands protect the kidney from ischemia reperfusion injury. J Immunol. 2015;194:325–33.

    Article  CAS  PubMed  Google Scholar 

  96. Nossent AY, Bastiaansen AJ, Peters EA, de Vries MR, Aref Z, Welten SM, et al. CCR7-CCL19/CCL21 axis is essential for effective arteriogenesis in a murine model of hindlimb ischemia. J Am Heart Assoc. 2017;6:1.

    Article  Google Scholar 

  97. Latet SC, Hoymans VY, Van Herck PL, Vrints CJ. The cellular immune system in the post-myocardial infarction repair process. Int J Cardiol. 2015;179:240–7.

    Article  PubMed  Google Scholar 

  98. Zuidema MY, Zhang C. Ischemia/reperfusion injury: The role of immune cells. World journal of cardiology. 2010;2:325–32.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Hofmann U, Frantz S. Role of lymphocytes in myocardial injury, healing, and remodeling after myocardial infarction. Circ Res. 2015;116:354–67.

    Article  CAS  PubMed  Google Scholar 

  100. Stabile E, Kinnaird T, la Sala A, Hanson SK, Watkins C, Campia U, et al. CD8+ T lymphocytes regulate the arteriogenic response to ischemia by infiltrating the site of collateral vessel development and recruiting CD4+ mononuclear cells through the expression of interleukin-16. Circulation. 2006;113:118–24.

    Article  PubMed  Google Scholar 

  101. van Weel V, Toes RE, Seghers L, Deckers MM, de Vries MR, Eilers PH, et al. Natural killer cells and CD4+ T-cells modulate collateral artery development. Arterioscler Thromb Vasc Biol. 2007;27:2310–8.

    Article  PubMed  CAS  Google Scholar 

  102. Beneke A, Guentsch A, Hillemann A, Zieseniss A, Swain L, Katschinski DM. Loss of PHD3 in myeloid cells dampens the inflammatory response and fibrosis after hind-limb ischemia. Cell Death Dis. 2017;8:e2976.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Oklu R, Albadawi H, Jones JE, Yoo HJ, Watkins MT. Reduced hind limb ischemia-reperfusion injury in Toll-like receptor-4 mutant mice is associated with decreased neutrophil extracellular traps. J Vasc Surg. 2013;58:1627–36.

    Article  PubMed  Google Scholar 

  104. Carbone F, Nencioni A, Mach F, Vuilleumier N, Montecucco F. Pathophysiological role of neutrophils in acute myocardial infarction. Thromb Haemost. 2013;110:501–14.

    Article  CAS  PubMed  Google Scholar 

  105. Stabile E, Burnett MS, Watkins C, Kinnaird T, Bachis A, la Sala A, et al. Impaired arteriogenic response to acute hindlimb ischemia in CD4-knockout mice. Circulation. 2003;108:205–10.

    Article  PubMed  Google Scholar 

  106. Hofmann U, Beyersdorf N, Weirather J, Podolskaya A, Bauersachs J, Ertl G, et al. Activation of CD4+ T lymphocytes improves wound healing and survival after experimental myocardial infarction in mice. Circulation. 2012;125:1652–63.

    Article  CAS  PubMed  Google Scholar 

  107. Simons KH, Aref Z, Peters HAB, Welten SP, Nossent AY, Jukema JW, et al. The role of CD27-CD70-mediated T cell co-stimulation in vasculogenesis, arteriogenesis and angiogenesis. Int J Cardiol. 2018;260:184–90.

    Article  CAS  PubMed  Google Scholar 

  108. Hellingman AA, Zwaginga JJ, van Beem RT, Hamming JF, Fibbe WE, Quax PH, et al. T-cell-pre-stimulated monocytes promote neovascularisation in a murine hind limb ischaemia model. Eur J Vasc Endovasc Surg. 2011;41:418–28.

    Article  CAS  PubMed  Google Scholar 

  109. Niiyama H, Kai H, Yamamoto T, Shimada T, Sasaki K, Murohara T, et al. Roles of endogenous monocyte chemoattractant protein-1 in ischemia-induced neovascularization. J Am Coll Cardiol. 2004;44:661–6.

    Article  CAS  PubMed  Google Scholar 

  110. Choe MA, Kim KH, An GJ, Lee KS, Heitkemper M. Hindlimb muscle atrophy occurs from peripheral nerve damage in a rat neuropathic pain model. Biol Res Nurs. 2011;13:44–54.

    Article  PubMed  Google Scholar 

  111. Tsuji N, Yamashita S, Sugawara Y, Kobayashi E. Effect of prolonged ischaemic time on muscular atrophy and regenerating nerve fibres in transplantation of the rat hind limb. J Plast Surg Hand Surg. 2012;46:217–21.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Ucuzian AA, Gassman AA, East AT, Greisler HP. Molecular mediators of angiogenesis. J Burn Care Res. 2010;31:158–75.

    Article  PubMed  Google Scholar 

  113. Jung S, Panchalingam KM, Wuerth RD, Rosenberg L, Behie LA. Large-scale production of human mesenchymal stem cells for clinical applications. Biotechnol Appl Biochem. 2012;59:106–20.

    Article  CAS  PubMed  Google Scholar 

  114. Eseonu OI, De Bari C. Homing of mesenchymal stem cells: mechanistic or stochastic? Implications for targeted delivery in arthritis. Rheumatology (Oxford). 2015;54:210–8.

    Article  Google Scholar 

  115. De Becker A, Riet IV. Homing and migration of mesenchymal stromal cells: how to improve the efficacy of cell therapy? World journal of stem cells. 2016;8:73–87.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Ullah M, Liu DD, Thakor AS. Mesenchymal stromal cell homing: mechanisms and strategies for improvement. IScience. 2019;15:421–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Zachar L, Bacenkova D, Rosocha J. Activation, homing, and role of the mesenchymal stem cells in the inflammatory environment. J Inflam Res. 2016;9:231–40.

    Article  CAS  Google Scholar 

  118. Wynn RF, Hart CA, Corradi-Perini C, O’Neill L, Evans CA, Wraith JE, et al. A small proportion of mesenchymal stem cells strongly expresses functionally active CXCR4 receptor capable of promoting migration to bone marrow. Blood. 2004;104:2643–5.

    Article  CAS  PubMed  Google Scholar 

  119. Ryu CH, Park SA, Kim SM, Lim JY, Jeong CH, Jun JA, et al. Migration of human umbilical cord blood mesenchymal stem cells mediated by stromal cell-derived factor-1/CXCR4 axis via Akt, ERK, and p38 signal transduction pathways. Biochem Biophys Res Commun. 2010;398:105–10.

    Article  CAS  PubMed  Google Scholar 

  120. Park SA, Ryu CH, Kim SM, Lim JY, Park SI, Jeong CH, et al. CXCR4-transfected human umbilical cord blood-derived mesenchymal stem cells exhibit enhanced migratory capacity toward gliomas. Int J Oncol. 2011;38:97–103.

    CAS  PubMed  Google Scholar 

  121. Marquez-Curtis LA, Janowska-Wieczorek A. Enhancing the migration ability of mesenchymal stromal cells by targeting the SDF-1/CXCR4 axis. Biomed Res Int. 2013;2013:561098.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  122. Steingen C, Brenig F, Baumgartner L, Schmidt J, Schmidt A, Bloch W. Characterization of key mechanisms in transmigration and invasion of mesenchymal stem cells. J Mol Cell Cardiol. 2008;44:1072–84.

    Article  CAS  PubMed  Google Scholar 

  123. Bayo J, Real A, Fiore EJ, Malvicini M, Sganga L, Bolontrade M, et al. IL-8, GRO and MCP-1 produced by hepatocellular carcinoma microenvironment determine the migratory capacity of human bone marrow-derived mesenchymal stromal cells without affecting tumor aggressiveness. Oncotarget. 2017;8:80235–48.

    Article  PubMed  Google Scholar 

  124. Bi LK, Zhou N, Liu C, Lu FD, Lin TX, Xuan XJ, et al. Kidney cancer cells secrete IL-8 to activate Akt and promote migration of mesenchymal stem cells. Urol Oncol. 2014;32:607–12.

    Article  CAS  PubMed  Google Scholar 

  125. Ponte AL, Marais E, Gallay N, Langonné A, Delorme B, Hérault O, et al. The in vitro migration capacity of human bone marrow mesenchymal stem cells: comparison of chemokine and growth factor chemotactic activities. Stem cells. 2007;25:1737–45.

    Article  CAS  PubMed  Google Scholar 

  126. Pfützner A, Schipper D, Pansky A, Kleinfeld C, Roitzheim B, Tobiasch E. Mesenchymal stem cell differentiation into adipocytes is equally induced by insulin and proinsulin in vitro. Int J Stem Cells. 2017;10:154–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Szaraz P, Gratch YS, Iqbal F, Librach CL. In vitro differentiation of human mesenchymal stem cells into functional cardiomyocyte-like cells. J Vis Experim JoVE. 2017;126:1.

    Google Scholar 

  128. Shivakumar SB, Lee HJ, Son YB, Bharti D, Ock SA, Lee SL, et al. In vitro differentiation of single donor derived human dental mesenchymal stem cells into pancreatic β cell-like cells. Bioscience Rep. 2019;39:1.

    Article  Google Scholar 

  129. Urrutia DN, Caviedes P, Mardones R, Minguell JJ, Vega-Letter AM, Jofre CM. Comparative study of the neural differentiation capacity of mesenchymal stromal cells from different tissue sources: An approach for their use in neural regeneration therapies. PLoS One. 2019;14:e0213032.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Dos Santos A, Balayan A, Funderburgh ML, Ngo J, Funderburgh JL, Deng SX. Differentiation capacity of human mesenchymal stem cells into keratocyte lineage. Invest Ophthalmol Vis Sci. 2019;60:3013–23.

    Article  PubMed  CAS  Google Scholar 

  131. Espagnolle N, Guilloton F, Deschaseaux F, Gadelorge M, Sensebe L, Bourin P. CD146 expression on mesenchymal stem cells is associated with their vascular smooth muscle commitment. J Cell Mol Med. 2014;18:104–14.

    Article  CAS  PubMed  Google Scholar 

  132. Gu W, Hong X, Le Bras A, Nowak WN, Issa Bhaloo S, Deng J, et al. Smooth muscle cells differentiated from mesenchymal stem cells are regulated by microRNAs and suitable for vascular tissue grafts. J Biol Chem. 2018;293:8089–102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Yu Q, Liu L, Lin J, Wang Y, Xuan X, Guo Y, et al. SDF-1alpha/CXCR4 axis mediates the migration of mesenchymal stem cells to the hypoxic-ischemic brain lesion in a rat model. Cell J. 2015;16:440–7.

    PubMed  PubMed Central  Google Scholar 

  134. Bang OY, Jin KS, Hwang MN, Kang HY, Kim BJ, Lee SJ, et al. The Effect of CXCR4 overexpression on mesenchymal stem cell transplantation in ischemic stroke. Cell Med. 2012;4:65–76.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Wu SZ, Li YL, Huang W, Cai WF, Liang J, Paul C, et al. Paracrine effect of CXCR4-overexpressing mesenchymal stem cells on ischemic heart injury. Cell Biochem Funct. 2017;35:113–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Rostovskaya M, Anastassiadis K. Differential expression of surface markers in mouse bone marrow mesenchymal stromal cell subpopulations with distinct lineage commitment. PLoS One. 2012;7:e51221.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Shahror RA, Ali AAA, Wu CC, Chiang YH, Chen KY. Enhanced Homing of Mesenchymal Stem Cells Overexpressing Fibroblast Growth Factor 21 to Injury Site in a Mouse Model of Traumatic Brain Injury. Int J Mol Sci. 2019;20:1.

    Article  CAS  Google Scholar 

  138. Bian X, Ma K, Zhang C, Fu X. Therapeutic angiogenesis using stem cell-derived extracellular vesicles: an emerging approach for treatment of ischemic diseases. Stem Cell Res Ther. 2019;10:158.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Beegle JR, Magner NL, Kalomoiris S, Harding A, Zhou P, Nacey C, et al. Preclinical evaluation of mesenchymal stem cells overexpressing VEGF to treat critical limb ischemia. Mol Ther Methods Clin Dev. 2016;3:16053.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  140. Arutyunyan I, Fatkhudinov T, Kananykhina E, Usman N, Elchaninov A, Makarov A, et al. Role of VEGF-A in angiogenesis promoted by umbilical cord-derived mesenchymal stromal/stem cells: in vitro study. Stem Cell Res Ther. 2016;7:46.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  141. Jeong CH, Kim SM, Lim JY, Ryu CH, Jun JA, Jeun SS. Mesenchymal stem cells expressing brain-derived neurotrophic factor enhance endogenous neurogenesis in an ischemic stroke model. Biomed Res Int. 2014;2014:129145.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Tobin MK, Stephen TKL, Lopez KL, Pergande MR, Bartholomew AM, Cologna SM, et al. Activated mesenchymal stem cells induce recovery following stroke via regulation of inflammation and oligodendrogenesis. J Am Heart Assoc. 2020;9:e013583.

    Article  PubMed  PubMed Central  Google Scholar 

  143. He H, Zeng Q, Huang G, Lin Y, Lin H, Liu W, et al. Bone marrow mesenchymal stem cell transplantation exerts neuroprotective effects following cerebral ischemia/reperfusion injury by inhibiting autophagy via the PI3K/Akt pathway. Brain Res. 2019;1707:124–32.

    Article  CAS  PubMed  Google Scholar 

  144. Zheng Z, Zhang L, Qu Y, Xiao G, Li S, Bao S, et al. Mesenchymal stem cells protect against hypoxia-ischemia brain damage by enhancing autophagy through brain derived neurotrophic factor/mammalin target of rapamycin signaling pathway. Stem Cells. 2018;36:1109–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Kasper G, Dankert N, Tuischer J, Hoeft M, Gaber T, Glaeser JD, et al. Mesenchymal stem cells regulate angiogenesis according to their mechanical environment. Stem Cells. 2007;25:903–10.

    Article  CAS  PubMed  Google Scholar 

  146. Hung SC, Pochampally RR, Chen SC, Hsu SC, Prockop DJ. Angiogenic effects of human multipotent stromal cell conditioned medium activate the PI3K-Akt pathway in hypoxic endothelial cells to inhibit apoptosis, increase survival, and stimulate angiogenesis. Stem Cells. 2007;25:2363–70.

    Article  CAS  PubMed  Google Scholar 

  147. Gonzalez-King H, Garcia NA, Ontoria-Oviedo I, Ciria M, Montero JA, Sepulveda P. Hypoxia inducible factor-1alpha potentiates jagged 1-mediated angiogenesis by mesenchymal stem cell-derived exosomes. Stem cells. 2017;35:1747–59.

    Article  CAS  PubMed  Google Scholar 

  148. Serra J, Alves CPA, Brito L, Monteiro GA, Cabral JMS, Prazeres DMF, et al. Engineering of human mesenchymal stem/stromal cells with vascular endothelial growth factor-encoding minicircles for angiogenic ex vivo gene therapy. Hum Gene Ther. 2019;30:316–29.

    Article  CAS  PubMed  Google Scholar 

  149. Arutyunyan IV, Kananykhina EY, Fatkhudinov T, El’chaninov AV, Makarov AV, Raimova E, et al. Angiogenic potential of multipotent stromal cells from the umbilical cord: an in vitro study. Bull Exp Biol Med. 2016;161:141–9.

    Article  CAS  PubMed  Google Scholar 

  150. Boareto M, Jolly MK, Ben-Jacob E, Onuchic JN. Jagged mediates differences in normal and tumor angiogenesis by affecting tip-stalk fate decision. Proc Natl Acad Sci U S A. 2015;112:E3836–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Munir H, Ward LSC, Sheriff L, Kemble S, Nayar S, Barone F, et al. Adipogenic differentiation of mesenchymal stem cells alters their immunomodulatory properties in a tissue-specific manner. Stem cells. 2017;35:1636–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Yang ZX, Han ZB, Ji YR, Wang YW, Liang L, Chi Y, et al. CD106 identifies a subpopulation of mesenchymal stem cells with unique immunomodulatory properties. PLoS One. 2013;8:e59354.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Bowles AC, Kouroupis D, Willman MA, Orfei CP, Agarwal A, Correa D. CD146+CD107a+ mesenchymal stem/stromal cells with signature attributes correlate to therapeutic potency as “first responders” to injury and inflammation. bioRxiv. 2019;2019:787176.

    Google Scholar 

  154. Anderson P, Carrillo-Gálvez AB, García-Pérez A, Cobo M, Martín F. CD105 (endoglin)-negative murine mesenchymal stromal cells define a new multipotent subpopulation with distinct differentiation and immunomodulatory capacities. PLoS One. 2013;8:e76979.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Pham LH, Vu NB, Van Pham P. The subpopulation of CD105 negative mesenchymal stem cells show strong immunomodulation capacity compared to CD105 positive mesenchymal stem cells. Biomed Res Ther. 2019;6:3131–40.

    Article  Google Scholar 

  156. Blazquez R, Sanchez-Margallo FM, de la Rosa O, Dalemans W, Alvarez V, Tarazona R, et al. Immunomodulatory potential of human adipose mesenchymal stem cells derived exosomes on in vitro stimulated T cells. Front Immunol. 2014;5:556.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  157. Khare D, Or R, Resnick I, Barkatz C, Almogi-Hazan O, Avni B. Mesenchymal stromal cell-derived exosomes affect mRNA expression and function of B-lymphocytes. Front Immunol. 2018;9:3053.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105:1815–22.

    Article  CAS  PubMed  Google Scholar 

  159. Zimmermann JA, Hettiaratchi MH, McDevitt TC. Enhanced immunosuppression of T cells by sustained presentation of bioactive interferon-γ within three-dimensional mesenchymal stem cell constructs. Stem Cells Transl Med. 2017;6:223–37.

    Article  CAS  PubMed  Google Scholar 

  160. Beegle J, Lakatos K, Kalomoiris S, Stewart H, Isseroff RR, Nolta JA, et al. Hypoxic preconditioning of mesenchymal stromal cells induces metabolic changes, enhances survival, and promotes cell retention in vivo. Stem cells. 2015;33:1818–28.

    Article  CAS  PubMed  Google Scholar 

  161. Ferro F, Spelat R, Shaw G, Duffy N, Islam MN, O’Shea PM, et al. Survival/adaptation of bone marrow-derived mesenchymal stem cells after long-term starvation through selective processes. Stem cells. 2019;37:813–27.

    Article  CAS  PubMed  Google Scholar 

  162. Hofmann M, Wollert KC, Meyer GP, Menke A, Arseniev L, Hertenstein B, et al. Monitoring of bone marrow cell homing into the infarcted human myocardium. Circulation. 2005;111:2198–202.

    Article  PubMed  Google Scholar 

  163. Mao AS, Özkale B, Shah NJ, Vining KH, Descombes T, Zhang L, et al. Programmable microencapsulation for enhanced mesenchymal stem cell persistence and immunomodulation. Proc Natl Acad Sci U S A. 2019;116:15392–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Bhakta S, Hong P, Koc O. The surface adhesion molecule CXCR4 stimulates mesenchymal stem cell migration to stromal cell-derived factor-1 in vitro but does not decrease apoptosis under serum deprivation. Cardiovasc Revascular Med: Includ Mol Interv. 2006;3:19–24.

    Article  Google Scholar 

  165. Won YW, Patel AN, Bull DA. Cell surface engineering to enhance mesenchymal stem cell migration toward an SDF-1 gradient. Biomaterials. 2014;35:5627–35.

    Article  CAS  PubMed  Google Scholar 

  166. Tebebi PA, Kim SJ, Williams RA, Milo B, Frenkel V, Burks SR, et al. Improving the therapeutic efficacy of mesenchymal stromal cells to restore perfusion in critical limb ischemia through pulsed focused ultrasound. Sci Rep. 2017;7:41550.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Yun WS, Choi JS, Ju HM, Kim MH, Choi SJ, Oh ES, et al. Enhanced homing technique of mesenchymal stem cells using iron oxide nanoparticles by magnetic attraction in olfactory-injured mouse models. Int J Mol Sci. 2018;19:1.

    Article  Google Scholar 

  168. Corradetti B, Taraballi F, Martinez JO, Minardi S, Basu N, Bauza G, et al. Hyaluronic acid coatings as a simple and efficient approach to improve MSC homing toward the site of inflammation. Sci Rep. 2017;7:7991.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  169. Zhidkova OV, Andreeva ER, Buravkova LB. Endothelial cells modulate differentiation potential and mobility of mesenchymal stromal cells. Bull Exp Biol Med. 2018;165:127–31.

    Article  CAS  PubMed  Google Scholar 

  170. Saini U, Gumina RJ, Wolfe B, Kuppusamy ML, Kuppusamy P, Boudoulas KD. Preconditioning mesenchymal stem cells with caspase inhibition and hyperoxia prior to hypoxia exposure increases cell proliferation. J Cell Biochem. 2013;114:2612–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Sivanathan KN, Rojas-Canales D, Grey ST, Gronthos S, Coates PT. Transcriptome profiling of IL-17A preactivated mesenchymal stem cells: a comparative study to unmodified and IFN-γ modified mesenchymal stem cells. Stem Cells Int. 2017;2017:1025820.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  172. Fan H, Zhao G, Liu L, Liu F, Gong W, Liu X, et al. Pre-treatment with IL-1β enhances the efficacy of MSC transplantation in DSS-induced colitis. Cell Mol Immunol. 2012;9:473–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Pourgholaminejad A, Aghdami N, Baharvand H, Moazzeni SM. The effect of pro-inflammatory cytokines on immunophenotype, differentiation capacity and immunomodulatory functions of human mesenchymal stem cells. Cytokine. 2016;85:51–60.

    Article  CAS  PubMed  Google Scholar 

  174. Zhang K, Chen X, Li H, Feng G, Nie Y, Wei Y, et al. A nitric oxide-releasing hydrogel for enhancing the therapeutic effects of mesenchymal stem cell therapy for hindlimb ischemia. Acta Biomater. 2020;113:289–304.

    Article  CAS  PubMed  Google Scholar 

  175. Kaushik K, Das A. Cycloxygenase-2 inhibition potentiates trans-differentiation of Wharton’s jelly-mesenchymal stromal cells into endothelial cells: transplantation enhances neovascularization-mediated wound repair. Cytotherapy. 2019;21:260–73.

    Article  CAS  PubMed  Google Scholar 

  176. Liu C, Tsai AL, Li PC, Huang CW, Wu CC. Endothelial differentiation of bone marrow mesenchyme stem cells applicable to hypoxia and increased migration through Akt and NFκB signals. Stem Cell Res Ther. 2017;8:29.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  177. Nguyen HT, Van Pham P. Conversion of human adipose derived stem cells into endothelial progenitor cells. Progress Stem Cell. 2017;4:229–40.

    Google Scholar 

  178. Toma C, Pittenger MF, Cahill KS, Byrne BJ, Kessler PD. Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation. 2002;105:93–8.

    Article  PubMed  Google Scholar 

  179. McGinley LM, McMahon J, Stocca A, Duffy A, Flynn A, O’Toole D, et al. Mesenchymal stem cell survival in the infarcted heart is enhanced by lentivirus vector-mediated heat shock protein 27 expression. Hum Gene Ther. 2013;24:840–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Dorland YL, Cornelissen AS, Kuijk C, Tol S, Hoogenboezem M, van Buul JD, et al. Nuclear shape, protrusive behaviour and in vivo retention of human bone marrow mesenchymal stromal cells is controlled by Lamin-A/C expression. Sci Rep. 2019;9:14401.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  181. Drela K, Stanaszek L, Nowakowski A, Kuczynska Z, Lukomska B. Experimental strategies of mesenchymal stem cell propagation: adverse events and potential risk of functional changes. Stem Cells Int. 2019;2019:7012692.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  182. Zhou Y, Singh AK, Hoyt RF Jr, Wang S, Yu Z, Hunt T, et al. Regulatory T cells enhance mesenchymal stem cell survival and proliferation following autologous cotransplantation in ischemic myocardium. J Thorac Cardiovasc Surg. 2014;148:1131–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Silva GV, Litovsky S, Assad JA, Sousa AL, Martin BJ, Vela D, et al. Mesenchymal stem cells differentiate into an endothelial phenotype, enhance vascular density, and improve heart function in a canine chronic ischemia model. Circulation. 2005;111:150–6.

    Article  CAS  PubMed  Google Scholar 

  184. Oswald J, Boxberger S, Jørgensen B, Feldmann S, Ehninger G, Bornhäuser M, et al. Mesenchymal stem cells can be differentiated into endothelial cells in vitro. Stem cells. 2004;22:377–84.

    Article  PubMed  Google Scholar 

  185. Crisan M. Transition of mesenchymal stem/stromal cells to endothelial cells. Stem Cell Res Ther. 2013;4:95.

    Article  PubMed  PubMed Central  Google Scholar 

  186. Alaminos M, Pérez-Köhler B, Garzón I, García-Honduvilla N, Romero B, Campos A, et al. Transdifferentiation potentiality of human Wharton’s jelly stem cells towards vascular endothelial cells. J Cell Physiol. 2010;223:640–7.

    CAS  PubMed  Google Scholar 

  187. Dao TT-T, Vu NB, Phi LT, Le HT-N, Phan NK, Van Pham P. Human adipose-derived mesenchymal stem cell could participate in angiogenesis in a mouse model of acute hindlimb ischemia. Biomed Res Ther. 2016;3:770–9.

    Article  Google Scholar 

  188. Corotchi MC, Popa MA, Remes A, Sima LE, Gussi I, Lupu PM. Isolation method and xeno-free culture conditions influence multipotent differentiation capacity of human Wharton’s jelly-derived mesenchymal stem cells. Stem Cell Research Ther. 2013;4:81.

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  190. Wang Y, Huang J, Gong L, Yu D, An C, Bunpetch V, et al. The Plasticity of Mesenchymal Stem Cells in Regulating Surface HLA-I. iScience. 2019;15:66–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Nauta AJ, Westerhuis G, Kruisselbrink AB, Lurvink EG, Willemze R, Fibbe WE. Donor-derived mesenchymal stem cells are immunogenic in an allogeneic host and stimulate donor graft rejection in a nonmyeloablative setting. Blood. 2006;108:2114–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Isakova IA, Lanclos C, Bruhn J, Kuroda MJ, Baker KC, Krishnappa V, et al. Allo-reactivity of mesenchymal stem cells in rhesus macaques is dose and haplotype dependent and limits durable cell engraftment in vivo. PLoS One. 2014;9:e87238.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  193. Ankrum JA, Ong JF, Karp JM. Mesenchymal stem cells: immune evasive, not immune privileged. Nat Biotechnol. 2014;32:252–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Eliopoulos N, Stagg J, Lejeune L, Pommey S, Galipeau J. Allogeneic marrow stromal cells are immune rejected by MHC class I- and class II-mismatched recipient mice. Blood. 2005;106:4057–65.

    Article  CAS  PubMed  Google Scholar 

  195. Hoornaert CJ, Luyckx E, Reekmans K, Dhainaut M, Guglielmetti C, Le Blon D, et al. In vivo interleukin-13-primed macrophages contribute to reduced alloantigen-specific t cell activation and prolong immunological survival of allogeneic mesenchymal stem cell implants. Stem Cells. 2016;34:1971–84.

    Article  CAS  PubMed  Google Scholar 

  196. Herrera OB, Golshayan D, Tibbott R, Salcido Ochoa F, James MJ, Marelli-Berg FM, et al. A novel pathway of alloantigen presentation by dendritic cells. J Immunol. 2004;173:4828–37.

    Article  CAS  PubMed  Google Scholar 

  197. Amorin B, Alegretti AP, Valim V, Pezzi A, Laureano AM, da Silva MA, et al. Mesenchymal stem cell therapy and acute graft-versus-host disease: a review. Hum Cell. 2014;27:137–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Sato K, Ozaki K, Mori M, Muroi K, Ozawa K. Mesenchymal stromal cells for graft-versus-host disease: basic aspects and clinical outcomes. J Clin Exp Hematopathol. 2010;50:79-89.

    Article  Google Scholar 

  199. Godoy JAP, Paiva RMA, Souza AM, Kondo AT, Kutner JM, Okamoto OK. Clinical translation of mesenchymal stromal cell therapy for graft versus host disease. Front Cell Dev Biol. 2019;7:255.

    Article  PubMed  PubMed Central  Google Scholar 

  200. Paronis E, Katsimpoulas M, Kadoglou NPE, Provost C, Stasinopoulou M, Spyropoulos C, et al. Cilostazol mediates immune responses and affects angiogenesis during the acute phase of hind limb ischemia in a mouse model. J Cardiovasc Pharmacol Ther. 2020;25:273–85.

    Article  CAS  PubMed  Google Scholar 

  201. 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.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  202. Yu H, Lu K, Zhu J, Wang J. Stem cell therapy for ischemic heart diseases. Br Med Bull. 2017;121:135–54.

    Article  PubMed  CAS  Google Scholar 

  203. Haque N, Kasim NH, Rahman MT. Optimization of pre-transplantation conditions to enhance the efficacy of mesenchymal stem cells. Int J Biol Sci. 2015;11:324–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

All authors equally contributed in this work. All authors read and approved the final manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Phuc Van Pham.

Ethics declarations

Conflict of interests

The authors declare that they have no conflict of interest.

Ethical statement

There are no animal experiments carried out for this article.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Van Nguyen, TT., Vu, N.B. & Van Pham, P. Mesenchymal Stem Cell Transplantation for Ischemic Diseases: Mechanisms and Challenges. Tissue Eng Regen Med 18, 587–611 (2021). https://doi.org/10.1007/s13770-021-00334-3

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13770-021-00334-3

Keywords

Navigation