#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Mesenchymal stem cells: biology and potential in the treatment of patients with connective tissue disease


Authors: L. Formanová 1;  V. Vodičková 1;  I. Hromadníková 2;  R. Klubal 1
Authors‘ workplace: Medicínské centrum Praha 1;  Oddělení molekulární biologie a patologie buňky, 3. LF UK, Praha 2
Published in: Čes. Revmatol., 28, 2020, No. 1, p. 39-51.
Category: Review Article

Overview

Mesenchymal stem cells are a unique type of cells that have the ability of differentiation to another cell type and modulation of the immune system. Mesenchymal stem cells are easily obtainable from many tissues, they are able to differentiate in osteocytes, chondrocytes and adipocytes and capable to produce various immunomodulatory and immunoregulatory factors. Knowledge of the origin, life cycle and function is important for understanding of their mechanism of action in the treatment of multiple disorders, inclusive of autoimmune rheumatic diseases (i.e. systemic lupus erythematodes and rheumatoid arthritis). The aim of this review is to raise public awareness of the properties of mesenchymal stem cells and the possibilities of their potential exploitation in the treatment of connective tissue disorders.

Keywords:

immunomodulation – mesenchymal stem cells – rheumatoid arthritis – reparation – lupus erythematodes


Sources

1. Tuch BE. Stem cells – a clinical update. Aust Fam Physician 2006; 35: 719–721.

2. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006; 8: 315–317.

3. Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells Dayt 2007; 25: 2739–2749.

4. Lee WS, Kim HJ, Kim KI, Kim GB, Jin W. Intra-articular injection of autologous adipose tissue-derived mesenchymal stem cells for the treatment of knee osteoarthritis: a phase IIb, randomized, placebo-controlled clinical trial. Stem Cells Transl Med 2019; 8: 504–511.

5. Cho J, D’Antuono M, Glicksman M, Wang J, Jonklaas J. A review of clinical trials: mesenchymal stem cell transplant therapy in type 1 and type 2 diabetes mellitus. Am J Stem Cells 2018; 7: 82–93.

6. Álvaro-Gracia JM, Jover JA, García-Vicuña R, Carreño L, Alonso A, Marsal S, et al. Intravenous administration of expanded allogeneic adipose-derived mesenchymal stem cells in refractory rheumatoid arthritis (Cx611): results of a multicentre, dose escalation, randomised, single-blind, placebo-controlled phase Ib/IIa clinical trial. Ann Rheum Dis 2017; 76: 196–202.

7. Cuascut FX, Hutton GJ. Stem Cell-Based Therapies for Multiple Sclerosis: Current Perspectives Biomedicines 2019; 7(2): 26.

8. Sun L, Wang D, Liang J, Zhang H, Feng X, Wang H, et al. Umbilical cord mesenchymal stem cell transplantation in severe and refractory systemic lupus erythematosus. Arthritis Rheum 2010; 62: 2467–2475.

9. Kingery MT, Manjunath AK, Anil U, Strauss EJ. Bone marrow mesenchymal stem cell therapy and related bone marrow-derived orthobiologic therapeutics. Curr Rev Musculoskelet Med 2019; 12(4): 451–459.

10. Williams JT, Southerland SS, Souza J, Calcutt AF, Cartledge RG. Cells isolated from adult human skeletal muscle capable of differentiating into multiple mesodermal phenotypes. Am Surg 1999; 65: 22–26.

11. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng 2011; 7: 211–228.

12. Gancheva MR, Kremer KL, Gronthos S, Koblar SA. Using dental pulp stem cells for stroke therapy. Front Neurol 2019; 10: 422.

13. Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol 2000; 109: 235–242.

14. Piskorska-Jasiulewicz MM, Witkowska-Zimny M. Perinatal sources of stem cells. Postepy Hig Med Dosw (Online) 2015; 69: 327–334.

15. 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®) Mesenchymal Stromal Cell committee position statement on nomenclature. Cytotherapy 2019; 21: 1019–1024.

16. Squillaro T, Peluso G, Galderisi U. Clinical trials with mesenchymal stem cells: an update. Cell Transplant 2016; 25: 829–848.

17. Ullah I, Subbarao RB, Rho GJ. Human mesenchymal stem cells – current trends and future prospective. Biosci Rep 2015; 35: e00191.

18. Li N, Long B, Han W, Yuan S, Wang K. MicroRNAs: important regulators of stem cells. Stem Cell Res Ther 2017; 8: 110.

19. Kern S, Eichler H, Stoeve J, Klüter H, Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells 2006; 24 : 1294–1301.

20. Yang YHK. Aging of mesenchymal stem cells: Implication in regenerative medicine. Regen Ther 2018; 9: 120–122.

21. 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: 163–173.

22. Ahmed ASI, Sheng MH, Wasnik S, Baylink DJ, Lau KHW. Effect of aging on stem cells. World J Exp Med 2017; 7: 1–10.

23. Alt EU, Senst C, Murthy SN, Slakey DP, Dupin CL, Chaffin AE, Kadowitz PJ, et al. Aging alters tissue resident mesenchymal stem cell properties. Stem Cell Res 2012; 8: 215–225.

24. Kuo TK, Hung SP, Chuang CH, Chen CT, Shih YR, Fang SC, Yang VW, et al. Stem cell therapy for liver disease: parameters governing the success of using bone marrow mesenchymal stem cells. Gastroenterology 2008; 134: 2111–2121.

25. Aurich H, Sgodda1 M, Kaltwaßer P, Vetter M, Weise A, Liehr T, Brulport M, et al. Hepatocyte differentiation of mesenchymal stem cells from human adipose tissue in vitro promotes hepatic integration in vivo. Gut 2009; 58: 570–581.

26. Konala VBR, Mamidi MK, Bhonde R, Das AK, Pochampally R, Pal R, et al. The current landscape of the mesenchymal stromal cell secretome: A new paradigm for cell-free regeneration. Cytotherapy 2016; 18: 13–24.

27. Bruno S, Deregibus MC, Camussi G. The secretome of mesenchymal stromal cells: Role of extracellular vesicles in immunomodulation. Immunol Lett 2015; 168: 154–158.

28. Kobolak J, Dinnyes A, Memic A, Khademhosseini A, Mobasheri A. Mesenchymal stem cells: Identification, phenotypic characterization, biological properties and potential for regenerative medicine through biomaterial micro-engineering of their niche. Methods 2016; 99: 62–68.

29. Hernanda PY, Pedroza-Gonzalez A, van der Laan LJ, Bröker ME, Hoogduijn MJ, Ijzermans JN, et al. Tumor promotion through the mesenchymal stem cell compartment in human hepatocellular carcinoma. Carcinogenesis 2013; 34: 2330–2340.

30. Dalal J, Gandy K, Domen J. Role of mesenchymal stem cell therapy in Crohn’s disease. Pediatr Res 2012; 71: 445–451.

31. Katuchova J, et al. Mesenchymal stem cells in the treatment of type 1 diabetes mellitus. Endocr Pathol 2015; 26: 95–103.

32. Simmons PJ, Torok-Storb B. CD34 expression by stromal precursors in normal human adult bone marrow. Blood 1991; 78: 2848–2853.

33. Stagg J, Pommey S, Eliopoulos N, Galipeau J. Interferon-gamma-stimulated marrow stromal cells: a new type of nonhematopoietic antigen-presenting cell. Blood 2006; 107: 2570–2577.

34. Romieu-Mourez R, François M, Boivin MN, Stagg J, Galipeau J. Regulation of MHC class II expression and antigen processing in murine and human mesenchymal stromal cells by IFN-gamma, TGF-beta, and cell density. J Immunol 2007; 179: 1549–1558.

35. Galderisi U, Giordano A. The gap between the physiological and therapeutic roles of mesenchymal stem cells. Med Res Rev 2014; 34: 1100–1126.

36. Krebsbach PH, Kuznetsov SA, Bianco P, Robey PG. Bone marrow stromal cells: characterization and clinical application. Crit Rev Oral Biol Med Off Publ Am Assoc Oral Biol 1999; 10: 165–181.

37. Fraser JK, Wulur I, Alfonso Z, Hedrick MH. Fat tissue: an underappreciated source of stem cells for biotechnology. Trends Biotechnol 2006; 24: 150–154.

38. Campagnoli C, Roberts IA, Kumar S, Bennett PR, Bellantuono I, Fisk NM. Identification of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver, and bone marrow. Blood 2001; 98: 2396–2402.

39. Nekanti U, Rao VB, Bahirvani AG, Jan M, Totey S, Ta M. Long-term expansion and pluripotent marker array analysis of Wharton’s jelly-derived mesenchymal stem cells. Stem Cells Dev 2010; 19: 117–130.

40. La Rocca G, Anzalone R, Corrao S, Magno F, Loria T, Lo Iacono M, et al. Isolation and characterization of Oct-4+/HLA-G+ mesenchymal stem cells from human umbilical cord matrix: differentiation potential and detection of new markers. Histochem Cell Biol 2009; 131: 267–282.

41. Rebuzzini P, Neri T, Mazzini G, Zuccotti M, Redi CA, Garagna S. Karyotype analysis of the euploid cell population of a mouse embryonic stem cell line revealed a high incidence of chromosome abnormalities that varied during culture. Cytogenet Genome Res 2008; 121: 18–24.

42. Nagamura-Inoue T, He H. Umbilical cord-derived mesenchymal stem cells: Their advantages and potential clinical utility. World J Stem Cells 2014; 6: 195–202.

43. Vangsness CT, Sternberg H, Harris L. Umbilical cord tissue offers the greatest number of harvestable mesenchymal stem cells for research and clinical application: a literature review of different harvest sites. Arthroscopy 2015; 31: 1836–1843.

44. Cardoso TC, Ferrari HF, Garcia AF, Novais JB, Silva-Frade C, Ferrarezi MC, et al. Isolation and characterization of Wharton’s jelly-derived multipotent mesenchymal stromal cells obtained from bovine umbilical cord and maintained in a defined serum-free three-dimensional system. BMC Biotechnol 2012; 12: 18.

45. Zhu Y, et al. Placental mesenchymal stem cells of fetal and maternal origins demonstrate different therapeutic potentials. Stem Cell Res Ther 2014; 5: 48.

46. Fukuchi Y, et al. Human placenta-derived cells have mesenchymal stem/progenitor cell potential. Stem Cells 2004; 22: 649–658.

47. Marcus AJ, Woodbury D. Fetal stem cells from extra-embryonic tissues: do not discard. J Cell Mol Med 2008; 12: 730–742.

48. Sagar R, Walther-Jallow L, David AL, Götherström C, Westgren M. Fetal mesenchymal stromal cells: an opportunity for prenatal cellular therapy. Curr Stem Cell Rep 2018; 4: 61–68.

49. Götherström C, Westgren M, Shaw SW, Aström E, Biswas A, Byers PH, Mattar CN, et al. Pre- and postnatal transplantation of fetal mesenchymal stem cells in osteogenesis imperfecta: a two-center experience. Stem Cells Transl Med 2014; 3: 255–264.

50. Hromadnikova I, Zlacka D, Hien Nguyen TT, Sedlackova L, Zejskova L, Sosna A. Fetal cells of mesenchymal origin in cultures derived from synovial tissue and skin of patients with rheumatoid arthritis. J Bone Spine Rev Rhum 2008; 75: 563–566.

51. O’Donoghue K, Chan J. Human fetal mesenchymal stem cells. Curr Stem Cell Res Ther 2006; 1: 371–386.

52. Murray IR, West CC, Hardy WR, James AW, Park TS, Nguyen A, Tawonsawatruk T. Natural history of mesenchymal stem cells, from vessel walls to culture vessels. Cell Mol Life Sci CMLS 2014; 71: 1353–1374.

53. Chen Q, Shou P, Zheng C, Jiang M, Cao G, Yang Q, et al. Fate decision of mesenchymal stem cells: adipocytes or osteoblasts? Cell Death Differ 2016; 23: 1128–1139.

54. Patel SA, Sherman L, Munoz J, Rameshwar P. Immunological properties of mesenchymal stem cells and clinical implications. Arch Immunol Ther Exp (Warsz) 2008; 56: 1–8.

55. Ra JC, et al. Safety of intravenous infusion of human adipose tissue-derived mesenchymal stem cells in animals and humans. Stem Cells Dev 2011; 20: 1297–1308.

56. Home – ClinicalTrials.gov. https://clinicaltrials.gov/

57. Luque-Campos N, Contreras-López RA, Jose Paredes-Martínez M, Torres MJ, Bahraoui S, Wei M, et al. Mesenchymal stem cells improve rheumatoid Arthritis progression by controlling memory T cell response. Front Immunol 2019; 10: 798.

58. Chen W, Yu Y, Ma J, Olsen N, Lin J. Mesenchymal stem cells in primary Sjögren’s syndrome: prospective and challenges. Stem Cells Int 2018: 4357865.

59. Deng Y, 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.

60. Melief SM, et al. Multipotent stromal cells induce human regulatory T cells through a novel pathway involving skewing of monocytes toward anti-inflammatory macrophages. Stem Cells 2013; 31: 1980–1991.

61. Zheng G, Huang R, Qiu G, Ge M, Wang J, Shu Q, et al. Mesenchymal stromal cell-derived extracellular vesicles: regenerative and immunomodulatory effects and potential applications in sepsis. Cell Tissue Res 2018; 374: 1–15.

62. Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 2005; 105: 1815–1822.

63. 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: 880–891.

64. 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: 1312–1320.

65. Gieseke F, et al. Human multipotent mesenchymal stromal cells use galectin-1 to inhibit immune effector cells. Blood 2010; 116: 3770–3779.

66. Weiss ARR, Dahlke MH. Immunomodulation by mesenchymal stem cells (MSCs): mechanisms of action of living, apoptotic, and dead MSCs. Front Immunol 2019; 10: 1191.

67. Sato K, et al. Nitric oxide plays a critical role in suppression of T-cell proliferation by mesenchymal stem cells. Blood 2007; 109: 228–234.

68. 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: 1327–1333.

69. Centeno CJ, Busse D, Kisiday J, Keohan C, Freeman M, Karli D. Increased knee cartilage volume in degenerative joint disease using percutaneously implanted, autologous mesenchymal stem cells. Pain Physician 2008; 11: 343–353.

70. van Buul GM, Siebelt M, Leijs MJ, Bos PK, Waarsing JH, Kops N, et al. Mesenchymal stem cells reduce pain but not degenerative changes in a mono-iodoacetate rat model of osteoarthritis. J Orthop Res Off Publ Orthop Res Soc 2014; 32: 1167–1174.

71. Freitag J, Bates D, Boyd R, Shah K, Barnard A, Huguenin L, et al. Mesenchymal stem cell therapy in the treatment of osteoarthritis: reparative pathways, safety and efficacy – a review. BMC Musculoskelet Disord 2016; 17: 230.

72. Shah K. Drury T, Roic I, Hansen P, Malin M, Boyd R, et al. Outcome of allogeneic adult stem cell therapy in dogs suffering from osteoarthritis and other joint defects. Stem Cells Int 2018; 7309201.

73. Roos EM, Roos HP, Lohmander LS, Ekdahl C, Beynnon BD. Knee Injury and Osteoarthritis Outcome Score (KOOS) – development of a self-administered outcome measure. J Orthop Sports Phys Ther 1998; 28: 88–96.

74. Bellamy N, Buchanan WW, Goldsmith CH, Campbell J, Stitt LW. Validation study of WOMAC: a health status instrument for measuring clinically important patient relevant outcomes to antirheumatic drug therapy in patients with osteoarthritis of the hip or knee. J Rheumatol 1988; 15: 1833–1840.

75. Kellgren JH, Lawrence JS. Radiological assessment of osteo-arthrosis. Ann Rheum Dis 1957; 16: 494–502.

76. Bastos R, et al. Intra-articular injection of culture-expanded mesenchymal stem cells with or without addition of platelet-rich plasma is effective in decreasing pain and symptoms in knee osteoarthritis: a controlled, double-blind clinical trial. Knee Surg Sports Traumatol Arthrosc 2019 [Epub ahead of print].

77. Shah N. Increasing knee range of motion using a unique sustained method. North Am J Sports Phys Ther NAJSPT 2008; 3: 110–113.

78. Ansboro S, Roelofs AJ, de Bari C. Mesenchymal stem cells for the management of rheumatoid arthritis: immune modulation, repair or both? Curr Opin Rheumatol 2017; 29: 201–207.

79. Liu R, Li X, Zhang Z, Zhou M, Sun Y, Su D, et al. Allogeneic mesenchymal stem cells inhibited T follicular helper cell generation in rheumatoid arthritis. Sci Rep 2015; 5: 12777.

80. Vos K, Visser H, Schreuder GM, de Vries RR, Zwinderman AH, Breedveld FC, et al. Human leukocyte antigen-DQ and DR polymorphisms predict rheumatoid arthritis outcome better than DR alone. Hum Immunol 2001; 62: 1217–1225.

81. Wu J, et al. Association of HLA-DQB1 polymorphisms with rheumatoid arthritis: a meta-analysis. Postgrad Med J 2017; 93: 618–625.

82. Alamanos Y, Drosos AA. Epidemiology of adult rheumatoid arthritis. Autoimmun Rev 2005; 4: 130–136.

83. Kallberg H, Padyukov L, Plenge RM, Ronnelid J, Gregersen PK, van der Helm-van Mil AH, et al. Gene-gene and gene-environment interactions involving HLA-DRB1, PTPN22, and smoking in two subsets of rheumatoid arthritis. Am J Hum Genet 2007; 80: 867–875.

84. McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med 2011; 365: 2205–2219.

85. Wang L, Wang L, Cong X, Liu G, Zhou J, Bai B, et al. Human umbilical cord mesenchymal stem cell therapy for patients with active rheumatoid arthritis: safety and efficacy. Stem Cells Dev 2013; 22: 3192–3202.

86. Ranganath VK, Khanna D, Paulus HE. ACR remission criteria and response criteria. Clin Exp Rheumatol 2006; 24(6 Suppl 43): S14–21.

87. Ali A, et al. Systemic Lupus Erythematosus: An Overview of the Disease Pathology and Its Management. Cureus 2018; 10(9): e3288.

88. Fortuna G, Brennan MT. Systemic lupus erythematosus: epidemiology, pathophysiology, manifestations, and management. Dent Clin North Am 2013; 57: 631–655.

89. Moser KL, Kelly JA, Lessard CJ, Harley JB. Recent insights into the genetic basis of systemic lupus erythematosus. Genes Immun 2009; 10: 373–379.

90. Graham RR, Hom G, Ortmann W, Behrens TW. Review of recent genome-wide association scans in lupus. J Intern Med 2009; 265: 680–688.

91. Bombardier C, Gladman DD, Urowitz MB, Caron D, Chang CH. Derivation of the SLEDAI. A disease activity index for lupus patients. The Committee on Prognosis Studies in SLE. Arthritis Rheum 1992; 35: 630–640.

92. Wang D, Akiyama K, Zhang H, Yamaza T, Li X, Feng X, Wang H, et al. Double allogenic mesenchymal stem cells transplantations could not enhance therapeutic effect compared with single transplantation in systemic lupus erythematosus. Clin Dev Immunol 2012; 2012: 273291.

93. Rendon A, Schäkel K. Psoriasis pathogenesis and treatment. Int J Mol Sci 2019; 20: 1475.

94. Indhumathi S, Rajappa M, Chandrashekar L, Anantha-narayanan PH, Thappa DM, Negi V. T helper-2 cytokine/regulatory T-cell gene polymorphisms and their relation with risk of psoriasis in a South Indian Tamil cohort. Hum Immunol 2017; 78: 209–215.

95. Osmola-Mańkowska A, Teresiak-Mikołajczak E, Skrzypczak-Zielińska M, Adamski Z. Genetic polymorphism in psoriasis and its meaning for the treatment efficacy in the future. Postepy Dermatol Alergol 2018; 35: 331–337.

96. Elder JT, Nair RP, Guo SW, Henseler T, Christophers E, Voorhees JJ. The genetics of psoriasis. Arch Dermatol 1994; 130: 216–224.

97. Alshobaili HA, Shahzad M, Al-Marshood A, Khalil AS, Settin A, Barrimah I. Genetic background of psoriasis. Int J Health Sci 2010; 4: 23–29.

98. Indhumathi S, Rajappa M, Chandrashekar L, Anantha-narayanan PH, Thappa DM, Negi VS. TNFAIP3 and TNIP1 polymorphisms confer psoriasis risk in South Indian Tamils. Br J Biomed Sci 2015; 72: 168–173.

99. Seetharaman R, Mahmood A, Kshatriya P, Patel D, Srivastava A. Mesenchymal stem cell conditioned media ameliorate psoriasis vulgaris: a case study. Case Rep Dermatol Med 2019; 8309103.

100. Menter A, Gottlieb A, Feldman SR, van Voorhees AS, Leonardi CL, Gordon KB, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis: Section 1. Overview of psoriasis and guidelines of care for the treatment of psoriasis with biologics. J Am Acad Dermatol 2008; 58: 826–850.

101. Spuls PI, Lecluse LL, Poulsen ML, Bos JD, Stern RS, Nijsten T. How good are clinical severity and outcome measures for psoriasis?: quantitative evaluation in a systematic review. J Invest Dermatol 2010; 130: 933–943.

102. Beckermann BM, Kallifatidis G, Groth A, Frommhold D, Apel A, Mattern J, et al. VEGF expression by mesenchymal stem cells contributes to angiogenesis in pancreatic carcinoma. Br J Cancer 2008; 99: 622–631.

103. Brennen WN, Nguyen H, Dalrymple SL, Reppert-Gerber S, Kim J, Isaacs JT, et al. Assessing angiogenic responses induced by primary human prostate stromal cells in a three-dimensional fibrin matrix assay. Oncotarget 2016; 7: 71298–71308.

104. Dvorak HF. Tumors: wounds that do not heal-a historical perspective with a focus on the fundamental roles of increased vascular permeability and clotting. Semin Thromb Hemost 2019; 45: 576–592.

105. Ridge SM, Sullivan FJ, Glynn SA. Mesenchymal stem cells: key players in cancer progression. Mol Cancer 2017; 16: 31.

106. Wu Y, Chen L, Scott PG, Tredget EE. Mesenchymal stem cells enhance wound healing through differentiation and angiogenesis. Stem Cells 2007; 25: 2648–2659.

107. Lee HY, Hong IS. Double-edged sword of mesenchymal stem cells: Cancer-promoting versus therapeutic potential. Cancer Sci 2017; 108: 1939–1946.

108. Vallabhaneni KC, Penfornis P, Dhule S, Guillonneau F, Adams KV, Mo YY, et al. Extracellular vesicles from bone marrow mesenchymal stem/stromal cells transport tumor regulatory microRNA, proteins, and metabolites. Oncotarget 2015; 6: 4953–4967.

109. Krueger TE, Thorek DLJ, Meeker AK, Isaacs JT, Brennen WN. Tumor-infiltrating mesenchymal stem cells: Drivers of the immunosuppressive tumor microenvironment in prostate cancer? The Prostate 2019; 79: 320–330.

110. Zhao S, Wehner R, Bornhäuser M, Wassmuth R, Bachmann M, Schmitz M, et al. Immunomodulatory properties of mesenchymal stromal cells and their therapeutic consequences for immune-mediated disorders. Stem Cells Dev 2010; 19: 607–614.

111. Rhee KJ, Lee JI, Eom YW. Mesenchymal stem cell-mediated effects of tumor support or suppression. Int J Mol Sci 2015; 16: 30015–30033.

112. Ho IA, Toh HC, Ng WH, Teo YL, Guo CM, Hui KM, et al. Human bone marrow-derived mesenchymal stem cells suppress human glioma growth through inhibition of angiogenesis. Stem Cells 2013; 31: 146–155.

113. Ohlsson LB, Varas L, Kjellman C, Edvardsen K, Lindvall M. Mesenchymal progenitor cell-mediated inhibition of tumor growth in vivo and in vitro in gelatin matrix. Exp Mol Pathol 2003; 75: 248–255.

114. Ramasamy R, Lam EW, Soeiro I, Tisato V, Bonnet D, Dazzi F. Mesenchymal stem cells inhibit proliferation and apoptosis of tumor cells: impact on in vivo tumor growth. Leukemia 2007; 21: 304–310.

115. Zhu Y, Sun Z, Han Q, Liao L, Wang J, Bian C, et al. Human mesenchymal stem cells inhibit cancer cell proliferation by secreting DKK-1. Leukemia 2009; 23: 925–933.

116. Sasaki A, Mizuno M, Mochizuki M, Sekiya I. Mesenchymal stem cells for cartilage regeneration in dogs. World J Stem Cells 2019; 11: 254–269.

117. Broeckx SY, et al. Equine allogeneic Chondrogenic induced mesenchymal stem cells are an effective treatment for degenerative joint disease in horses. Stem Cells Dev 2019; 28: 410–422.

118. Romero A, Barrachina L, Ranera B, Remacha AR, Moreno B, de Blas I, et al. Comparison of autologous bone marrow and adipose tissue derived mesenchymal stem cells, and platelet rich plasma, for treating surgically induced lesions of the equine superficial digital flexor tendon. Vet J 2017; 224: 76–84.

119. Taechangam N, Iyer SS, Walker NJ, Arzi B, Borjesson DL. Mechanisms utilized by feline adipose-derived mesenchymal stem cells to inhibit T lymphocyte proliferation. Stem Cell Res Ther 2019; 10: 188.

120. Quimby JM, Borjesson DL. Mesenchymal stem cell therapy in cats: Current knowledge and future potential. J Feline Med Surg 2018; 20: 208–216.

Labels
Dermatology & STDs Paediatric rheumatology Rheumatology
Login
Forgotten password

Enter the email address that you registered with. We will send you instructions on how to set a new password.

Login

Don‘t have an account?  Create new account

#ADS_BOTTOM_SCRIPTS#