Activated Endothelium Changes The Activity Of Multipotent Mesenchymal Stromal Cells During Physiological Hypoxia Or Short Hypoxic Stress In Vitro

Cover Page

Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription Access

Abstract

Multipotent mesenchymal stromal cells (MSCs) are used for supplemental therapy of ischemic and inflammatory diseases. After systemic administration, transmigration of MSCs to the target tissue is accompanied by interaction with activated endothelial cells (ECs) at the site of injury. In this study, we investigated the influence of TNF-α-activated ECs on the functions of MSCs under different levels of hypoxia. For this purpose, MSCs and TNF-α activated ECs were cocultured in a direct cell-to-cell setting for a short period of time. MSCs retained their stromal phenotype and multilineage differentiation potential after interaction with activated ECs. At the same time, changes in molecules involved in MSC-cell and MSC-extracellular matrix interaction were detected. The paracrine activity of MSCs and activated ECs after interaction was demonstrated by both upregulated transcription and increased levels of pleiotropic IL-6 and IL-8. Proteases/antiproteases profiles were also altered after interaction. These data suggest that short-term interaction of MSCs with activated ECs may play an important role in tissue repair and remodeling processes. In particular, it may promote the migratory phenotype of MSCs. In comparison to physiological hypoxia – 5% O2, acute hypoxic stress (0.1% O2, 24 h) attenuated the stimulatory effects of ECs on MSCs.

Full Text

Restricted Access

About the authors

O. V. Zhidkova

Institute of Biomedical Problems, Russian Academy of Sciences

Author for correspondence.
Email: flain-fish@yandex.ru
Russian Federation, Moscow

E. R. Andreeva

Institute of Biomedical Problems, Russian Academy of Sciences

Email: andreeva1564@gmail.com
Russian Federation, Moscow

L. B. Buravkova

Institute of Biomedical Problems, Russian Academy of Sciences

Email: buravkova@imbp.ru
Russian Federation, Moscow

References

  1. Caplan AI, Correa D (2011) The MSC: An Injury Drugstore. Cell Stem Cell 9: 11–15. https://doi.org/10.1016/j.stem.2011.06.008
  2. Squillaro T, Peluso G, Galderisi U (2016) Clinical Trials with Mesenchymal Stem Cells: An Update. Cell Transplant 25: 829–848. https://doi.org/10.3727/096368915X689622
  3. Galipeau J, Sensébé L (2018) Mesenchymal Stromal Cells: Clinical Challenges and Therapeutic Opportunities. Cell Stem Cell 22: 824–833. https://doi.org/10.1016/j.stem.2018.05.004
  4. Fernández-Garza LE, Barrera-Barrera SA, Barrera-Saldaña HA (2023) Mesenchymal Stem Cell Therapies Approved by Regulatory Agencies around the World. Pharmaceuticals 16: 1334. https://doi.org/10.3390/ph16091334
  5. Buravkova LB, Andreeva ER, Gogvadze V, Zhivotovsky B (2014) Mesenchymal stem cells and hypoxia: Where are we? Mitochondrion 19: 105–112. https://doi.org/10.1016/j.mito.2014.07.005
  6. Egger D, Lavrentieva A, Kugelmeier P, Kasper C (2022) Physiologic isolation and expansion of human mesenchymal stem/stromal cells for manufacturing of cell-based therapy products. Eng Life Sci 22: 361–372. https://doi.org/10.1002/elsc.202100097
  7. Choi J-W, Shin S, Lee CY, Lee J, Seo H-H, Lim S, Lee S, Kim I-K, Lee H-B, Kim SW, Hwang K-C (2017) Rapid Induction of Osteogenic Markers in Mesenchymal Stem Cells by Adipose-Derived Stromal Vascular Fraction Cells. Cell Physiol Biochem 44: 53–65. https://doi.org/10.1159/000484582
  8. Lavrentieva A, Majore I, Kasper C, Hass R (2010) Effects of hypoxic culture conditions on umbilical cord-derived human mesenchymal stem cells. Cell Commun Signal CCS 8: 18. https://doi.org/10.1186/1478-811X-8-18
  9. Moniz I, Ramalho-Santos J, Branco AF (2022) Differential Oxygen Exposure Modulates Mesenchymal Stem Cell Metabolism and Proliferation through mTOR Signaling. Int J Mol Sci 23: 3749. https://doi.org/10.3390/ijms23073749
  10. Rosová I, Dao M, Capoccia B, Link D, Nolta JA (2008) Hypoxic Preconditioning Results in Increased Motility and Improved Therapeutic Potential of Human Mesenchymal Stem Cells. Stem Cells 26: 2173–2182. https://doi.org/10.1634/stemcells.2007-1104
  11. Udartseva OO, Lobanova MV, Andreeva ER, Buravkov SV, Ogneva IV, Buravkova LB (2016) Acute Hypoxic Stress Affects Migration Machinery of Tissue O2-Adapted Adipose Stromal Cells. Stem Cells Int 2016: 1–16. https://doi.org/10.1155/2016/7260562
  12. Busletta C, Novo E, Valfrè Di Bonzo L, Povero D, Paternostro C, Ievolella M, Mareschi K, Ferrero I, Cannito S, Compagnone A, Bandino A, Colombatto S, Fagioli F, Parola M (2011) Dissection of the Biphasic Nature of Hypoxia-Induced Motogenic Action in Bone Marrow-Derived Human Mesenchymal Stem Cells. Stem Cells 29: 952–963. https://doi.org/10.1002/stem.642
  13. Wang Q, Li X, Wang Q, Xie J, Xie C, Fu X (2019) Heat shock pretreatment improves mesenchymal stem cell viability by heat shock proteins and autophagy to prevent cisplatin-induced granulosa cell apoptosis. Stem Cell Res Ther 10: 348. https://doi.org/10.1186/s13287-019-1425-4
  14. Peterson KM, Aly A, Lerman A, Lerman LO, Rodriguez-Porcel M (2011) Improved survival of mesenchymal stromal cell after hypoxia preconditioning: role of oxidative stress. Life Sci 88: 65–73. https://doi.org/10.1016/j.lfs.2010.10.023
  15. Andreeva ER, Lobanova MV, Udartseva OO, Buravkova LB (2014) Response of Adipose Tissue-Derived Stromal Cells in Tissue-Related O2 Microenvironment to Short-Term Hypoxic Stress. Cells Tissues Organs 200: 307–315. https://doi.org/10.1159/000438921
  16. Caplan AI (2007) Adult mesenchymal stem cells for tissue engineering versus regenerative medicine. J Cell Physiol 213: 341–347. https://doi.org/10.1002/jcp.21200
  17. Wei X, Yang X, Han Z, Qu F, Shao L, Shi Y (2013) Mesenchymal stem cells: a new trend for cell therapy. Acta Pharmacol Sin 34: 747–754. https://doi.org/10.1038/aps.2013.50
  18. Lu L, Xu A, Gao F, Tian C, Wang H, Zhang J, Xie Y, Liu P, Liu S, Yang C, Ye Z, Wu X (2022) Mesenchymal Stem Cell-Derived Exosomes as a Novel Strategy for the Treatment of Intervertebral Disc Degeneration. Front Cell Dev Biol 9: 770510. https://doi.org/10.3389/fcell.2021.770510
  19. Sanchez-Diaz M, Quiñones-Vico MI, Sanabria De La Torre R, Montero-Vílchez T, Sierra-Sánchez A, Molina-Leyva A, Arias-Santiago S (2021) Biodistribution of Mesenchymal Stromal Cells after Administration in Animal Models and Humans: A Systematic Review. J Clin Med 10: 2925. https://doi.org/10.3390/jcm10132925
  20. Becker AD, Riet IV (2016) Homing and migration of mesenchymal stromal cells: How to improve the efficacy of cell therapy? World J Stem Cells 8: 73. https://doi.org/10.4252/wjsc.v8.i3.73
  21. Szydlak R (2021) Biological, chemical and mechanical factors regulating migration and homing of mesenchymal stem cells. World J Stem Cells 13: 619–631. https://doi.org/10.4252/wjsc.v13.i6.619
  22. Leibacher J, Dauber K, Ehser S, Brixner V, Kollar K, Vogel A, Spohn G, Schäfer R, Seifried E, Henschler R (2017) Human mesenchymal stromal cells undergo apoptosis and fragmentation after intravenous application in immune-competent mice. Cytotherapy 19: 61–74. https://doi.org/10.1016/j.jcyt.2016.09.010
  23. Zachar L, Bačenková D, Rosocha J (2016) Activation, homing, and role of the mesenchymal stem cells in the inflammatory environment. J Inflamm Res 9: 231–240. https://doi.org/10.2147/JIR.S121994
  24. Nitzsche F, Müller C, Lukomska B, Jolkkonen J, Deten A, Boltze J (2017) Concise Review: MSC Adhesion Cascade—Insights into Homing and Transendothelial Migration. Stem Cells 35: 1446–1460. https://doi.org/10.1002/stem.2614
  25. Teo GSL, Ankrum JA, Martinelli R, Boetto SE, Simms K, Sciuto TE, Dvorak AM, Karp JM, Carman CV (2012) Mesenchymal Stem Cells Transmigrate Between and Directly Through Tumor Necrosis Factor-α-Activated Endothelial Cells Via Both Leukocyte-Like and Novel Mechanisms. Stem Cells 30: 2472–2486. https://doi.org/10.1002/stem.1198
  26. Guo Y-C, Chiu Y-H, Chen C-P, Wang H-S (2018) Interleukin-1β induces CXCR3-mediated chemotaxis to promote umbilical cord mesenchymal stem cell transendothelial migration. Stem Cell Res Ther 9: 281. https://doi.org/10.1186/s13287-018-1032-9
  27. Merfeld-Clauss S, Lupov IP, Lu H, Feng D, Compton-Craig P, March KL, Traktuev DO (2014) Adipose Stromal Cells Differentiate Along a Smooth Muscle Lineage Pathway Upon Endothelial Cell Contact via Induction of Activin A. Circ Res 115: 800–809. https://doi.org/10.1161/CIRCRESAHA.115.304026
  28. Lin C-H, Lilly B (2014) Endothelial Cells Direct Mesenchymal Stem Cells Toward a Smooth Muscle Cell Fate. Stem Cells Dev 23: 2581–2590. https://doi.org/10.1089/scd.2014.0163
  29. Li C, Wu X, Tong J, Yang X, Zhao J, Zheng Q, Zhao G, Ma Z (2015) Comparative analysis of human mesenchymal stem cells from bone marrow and adipose tissue under xeno-free conditions for cell therapy. Stem Cell Res Ther 6: 55. https://doi.org/10.1186/s13287-015-0066-5
  30. Buravkova LB, Grinakovskaya OS, Andreeva ER, Zhambalova AP, Kozionova MP (2009) Characteristics of human lipoaspirate-isolated mesenchymal stromal cells cultivated under lower oxygen tension. Cell Tissue Biol 3: 23–28. https://doi.org/10.1134/S1990519X09010039
  31. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini FC, Krause DS, Deans RJ, Keating A, Prockop DJ, Horwitz EM (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8: 315–317. https://doi.org/10.1080/14653240600855905
  32. Tondreau T, Meuleman N, Stamatopoulos B, De Bruyn C, Delforge A, Dejeneffe M, Martiat P, Bron D, Lagneaux L (2009) In vitro study of matrix metalloproteinase/tissue inhibitor of metalloproteinase production by mesenchymal stromal cells in response to inflammatory cytokines: the role of their migration in injured tissues. Cytotherapy 11: 559–569. https://doi.org/10.1080/14653240903051541
  33. Lozito TP, Jackson WM, Nesti LJ, Tuan RS (2014) Human mesenchymal stem cells generate a distinct pericellular zone of MMP activities via binding of MMPs and secretion of high levels of TIMPs. Matrix Biol 34: 132–143. https://doi.org/10.1016/j.matbio.2013.10.003
  34. Beloglazova IB, Zubkova ES, Tsokolaeva ZI, Stafeev YuS, Dergilev KV, Ratner EI, Shestakova MV, Sukhareva OYu, Parfenova EV, Menshikov MYu (2016) Regulatory Effects of Urokinase on Mesenchymal Stromal Cell Migration, Proliferation, and Matrix Metalloproteinase Secretion. Bull Exp Biol Med 161: 775–778. https://doi.org/10.1007/s10517-016-3507-y
  35. Wei L, Xu Y, Zhang L, Yang L, Zhao RC, Zhao D (2023) Mesenchymal Stem Cells Promote Wound Healing and Effects on Expression of Matrix Metalloproteinases-8 and 9 in the Wound Tissue of Diabetic Rats. Stem Cells Dev 32: 25–31. https://doi.org/10.1089/scd.2021.0218
  36. Lee JA, Kim BI, Jo CH, Choi CW, Kim E-K, Kim H-S, Yoon K-S, Choi J-H (2010) Mesenchymal stem-cell transplantation for hypoxic-ischemic brain injury in neonatal rat model. Pediatr Res 67: 42–46. https://doi.org/10.1203/PDR.0b013e3181bf594b
  37. Pogodina MV, Buravkova LB (2014) Expression of hypoxia-associated genes in multipotent mesenchymal stromal cells during long-term cultivation at low oxygen. Dokl Biol Sci 458: 310–312. https://doi.org/10.1134/S0012496614050056
  38. Pulido-Escribano V, Torrecillas-Baena B, Camacho-Cardenosa M, Dorado G, Gálvez-Moreno MÁ, Casado-Díaz A (2022) Role of hypoxia preconditioning in therapeutic potential of mesenchymal stem-cell-derived extracellular vesicles. World J Stem Cells 14: 453–472. https://doi.org/10.4252/wjsc.v14.i7.453
  39. Fontani F, Domazetovic V, Marcucci T, Vincenzini MT, Iantomasi T (2016) Tumor Necrosis Factor-Alpha Up-Regulates ICAM-1 Expression and Release in Intestinal Myofibroblasts by Redox-Dependent and -Independent Mechanisms: ICAM-1 Expression and Release in 18C o Cells. J Cell Biochem 117: 370–381. https://doi.org/10.1002/jcb.25279
  40. Li M, Van Esch BCAM, Wagenaar GTM, Garssen J, Folkerts G, Henricks PAJ (2018) Pro- and anti-inflammatory effects of short chain fatty acids on immune and endothelial cells. Eur J Pharmacol 831: 52–59. https://doi.org/10.1016/j.ejphar.2018.05.003
  41. Sigfrid LA, Cunningham JM, Beeharry N, Lortz S, Tiedge M, Lenzen S, Carlsson C, Green IC (2003) Cytokines and nitric oxide inhibit the enzyme activity of catalase but not its protein or mRNA expression in insulin-producing cells. J Mol Endocrinol 31: 509–518. https://doi.org/10.1677/jme.0.0310509
  42. Bauer G (2015) Increasing the endogenous NO level causes catalase inactivation and reactivation of intercellular apoptosis signaling specifically in tumor cells. Redox Biol 6: 353–371. https://doi.org/10.1016/j.redox.2015.07.017
  43. Mussbacher M, Salzmann M, Brostjan C, Hoesel B, Schoergenhofer C, Datler H, Hohensinner P, Basílio J, Petzelbauer P, Assinger A, Schmid JA (2019) Cell Type-Specific Roles of NF-κB Linking Inflammation and Thrombosis. Front Immunol 10: 85. https://doi.org/10.3389/fimmu.2019.00085
  44. Frith JE, Mills RJ, Hudson JE, Cooper-White JJ (2012) Tailored Integrin–Extracellular Matrix Interactions to Direct Human Mesenchymal Stem Cell Differentiation. Stem Cells Dev 21: 2442–2456. https://doi.org/10.1089/scd.2011.0615
  45. Steward AJ, Kelly DJ (2015) Mechanical regulation of mesenchymal stem cell differentiation. J Anat 227: 717–731. https://doi.org/10.1111/joa.12243
  46. Huttenlocher A, Horwitz AR (2011) Integrins in Cell Migration. Cold Spring Harb Perspect Biol 3: a005074–a005074. https://doi.org/10.1101/cshperspect.a005074
  47. Becerra-Bayona SM, Guiza-Arguello VR, Russell B, Höök M, Hahn MS (2018) Influence of collagen-based integrin α1 and α2 mediated signaling on human mesenchymal stem cell osteogenesis in three dimensional contexts. J Biomed Mater Res A 106: 2594–2604. https://doi.org/10.1002/jbm.a.36451
  48. Winning S, Splettstoesser F, Fandrey J, Frede S (2010) Acute Hypoxia Induces HIF-Independent Monocyte Adhesion to Endothelial Cells through Increased Intercellular Adhesion Molecule-1 Expression: The Role of Hypoxic Inhibition of Prolyl Hydroxylase Activity for the Induction of NF-κB. J Immunol 185: 1786–1793. https://doi.org/10.4049/jimmunol.0903244
  49. Thin Luu N, Mcgettrick HM, Buckley CD, Newsome PN, Ed Rainger G, Frampton J, Nash GB (2013) Crosstalk Between Mesenchymal Stem Cells and Endothelial Cells Leads to Downregulation of Cytokine-Induced Leukocyte Recruitment. Stem Cells 31: 2690–2702. https://doi.org/10.1002/stem.1511
  50. Bartaula-Brevik S (2017) Secretome of Mesenchymal Stem Cells Grown in Hypoxia Accelerates Wound Healing and Vessel Formation In Vitro. Int J Stem Cell Res Ther 4. https://doi.org/10.23937/2469-570X/1410045
  51. Hao Q, Wang L, Tang H (2009) Vascular endothelial growth factor induces protein kinase D-dependent production of proinflammatory cytokines in endothelial cells. Am J Physiol-Cell Physiol 296: C821–C827. https://doi.org/10.1152/ajpcell.00504.2008
  52. Guillamat-Prats R (2022) Role of Mesenchymal Stem/Stromal Cells in Coagulation. Int J Mol Sci 23: 10393. https://doi.org/10.3390/ijms231810393
  53. Lund LR, Green KA, Stoop AA, Ploug M, Almholt K, Lilla J, Nielsen BS, Christensen IJ, Craik CS, Werb Z, Danø K, Rømer J (2006) Plasminogen activation independent of uPA and tPA maintains wound healing in gene-deficient mice. EMBO J 25: 2686–2697. https://doi.org/10.1038/sj.emboj.7601173
  54. Rundhaug JE (2005) Matrix metalloproteinases and angiogenesis. J Cell Mol Med 9: 267–285. https://doi.org/10.1111/j.1582-4934.2005.tb00355.x
  55. McCarty SM, Percival SL (2013) Proteases and Delayed Wound Healing. Adv Wound Care 2: 438–447. https://doi.org/10.1089/wound.2012.0370
  56. Shalini S, Dorstyn L, Dawar S, Kumar S (2015) Old, new and emerging functions of caspases. Cell Death Differ 22: 526–539. https://doi.org/10.1038/cdd.2014.216
  57. Habič A, Novak M, Majc B, Lah Turnšek T, Breznik B (2021) Proteases Regulate Cancer Stem Cell Properties and Remodel Their Microenvironment. J Histochem Cytochem 69: 775–794. https://doi.org/10.1369/00221554211035192
  58. Brauer PR (2006) MMPs-Role in Cardiovascular Development and Disease. Front Biosci 11: 447. https://doi.org/10.2741/1810
  59. Kasper G, Glaeser JD, Geissler S, Ode A, Tuischer J, Matziolis G, Perka C, Duda GN (2007) Matrix Metalloprotease Activity Is an Essential Link Between Mechanical Stimulus and Mesenchymal Stem Cell Behavior. Stem Cells 25: 1985–1994. https://doi.org/10.1634/stemcells.2006-0676
  60. De Becker A, Van Hummelen P, Bakkus M, Vande Broek I, De Wever J, De Waele M, Van Riet I (2007) Migration of culture-expanded human mesenchymal stem cells through bone marrow endothelium is regulated by matrix metalloproteinase-2 and tissue inhibitor of metalloproteinase-3. Haematologica 92: 440–449. https://doi.org/10.3324/haematol.10475
  61. Ries C, Egea V, Karow M, Kolb H, Jochum M, Neth P (2007) MMP-2, MT1-MMP, and TIMP-2 are essential for the invasive capacity of human mesenchymal stem cells: differential regulation by inflammatory cytokines. Blood 109: 4055–4063. https://doi.org/10.1182/blood-2006-10-051060
  62. Tong B, Wan B, Wei Z, Wang T, Zhao P, Dou Y, Lv Z, Xia Y, Dai Y (2014) Role of cathepsin B in regulating migration and invasion of fibroblast-like synoviocytes into inflamed tissue from patients with rheumatoid arthritis. Clin Exp Immunol 177: 586–597. https://doi.org/10.1111/cei.12357
  63. Vangala G, Imhoff FM, Squires CML, Cridge AG, Baird SK (2019) Mesenchymal stem cell homing towards cancer cells is increased by enzyme activity of cathepsin D. Exp Cell Res 383: 111494. https://doi.org/10.1016/j.yexcr.2019.07.007
  64. Vidak E, Javoršek U, Vizovišek M, Turk B (2019) Cysteine Cathepsins and their Extracellular Roles: Shaping the Microenvironment. Cells 8: 264. https://doi.org/10.3390/cells8030264
  65. Park S, Huang H, Kwon W, Kim H, Park J, Han JE, Cho G, Han S, Sung Y, Ryoo ZY, Kim MO, Choi S (2021) Cathepsin A regulates pluripotency, proliferation and differentiation in mouse embryonic stem cells. Cell Biochem Funct 39: 67–76. https://doi.org/10.1002/cbf.3554
  66. Mott JD, Werb Z (2004) Regulation of matrix biology by matrix metalloproteinases. Curr Opin Cell Biol 16: 558–564. https://doi.org/10.1016/j.ceb.2004.07.010
  67. Stefansson S, Lawrence DA (1996) The serpin PAI-1 inhibits cell migration by blocking integrin αvβ3 binding to vitronectin. Nature 383: 441–443. https://doi.org/10.1038/383441a0
  68. Richard B, Pichon S, Arocas V, Venisse L, Berrou E, Bryckaert M, Jandrot Perrus M, Bouton MC (2006) The serpin protease nexin-1 regulates vascular smooth muscle cell adhesion, spreading, migration and response to thrombin. J Thromb Haemost 4: 322–328. https://doi.org/10.1111/j.1538-7836.2006.01710.x
  69. Provençal M, Michaud M, Beaulieu É, Ratel D, Rivard G-É, Gingras D, Béliveau R (2008) Tissue factor pathway inhibitor (TFPI) interferes with endothelial cell migration by inhibition of both the Erk pathway and focal adhesion proteins. Thromb Haemost 99: 576–585. https://doi.org/10.1160/TH07-10-0623
  70. Saller MM, Prall WC, Docheva D, Schönitzer V, Popov T, Anz D, Clausen-Schaumann H, Mutschler W, Volkmer E, Schieker M, Polzer H (2012) Increased stemness and migration of human mesenchymal stem cells in hypoxia is associated with altered integrin expression. Biochem Biophys Res Commun 423: 379–385. https://doi.org/10.1016/j.bbrc.2012.05.134
  71. Choi JH, Lee YB, Jung J, Hwang SG, Oh I-H, Kim GJ (2016) Hypoxia Inducible Factor-1 α Regulates the Migration of Bone Marrow Mesenchymal Stem Cells via Integrin α4. Stem Cells Int 2016: 1–11. https://doi.org/10.1155/2016/7932185
  72. Ren G, Roberts AI, Shi Y (2011) Adhesion molecules: Key players in mesenchymal stem cell-mediated immunosuppression. Cell Adhes Migr 5: 20–22. https://doi.org/10.4161/cam.5.1.13491
  73. Mai J, Virtue A, Shen J, Wang H, Yang X-F (2013) An evolving new paradigm: endothelial cells – conditional innate immune cells. J Hematol Oncol 6: 61. https://doi.org/10.1186/1756-8722-6-61

Supplementary files

Supplementary Files
Action
1. JATS XML
2. Fig. 1. Experimental scheme for evaluating the effects of interaction between mesenchymal stromal and endothelial cells.

Download (200KB)
3. Fig. 2. Characteristics of cultured MSCs with different O2 content. (a) – representative micrography of cultured MSCs at 5% O2, scale segment of 100 microns; (b) – characteristics of viability of MSCs using fluorescent dyes annexin/propidium iodide (Ann/PI); representative histograms are presented (lower left quadrant – population living cells, upper left quadrant – cells in a state of early apoptosis, upper right quadrant – cells in a state of late apoptosis, lower right quadrant – cells in a state of necrosis); (c) – production of ROS; (d) – expression of differentiation regulatory genes in MSCs, the multiplicity of changes in gene expression compared to the monoculture of MSCs at 5% O2 is presented; (e) – production of IL-6, IL-8 in MSCs. The graphs show the median, interquartile range, maximum and minimum values, n ≥ 4, * – p < 0.05 compared with the monoculture of MSC cultivated at 5% O2.

Download (263KB)
4. Fig. 3. Effect of TNF-α activation and O2 deprivation on EC at different O2 levels. (a) – intact and TNF-α activated EC after short–term cultivation at 5% O2, light microscopy, representative micrographs of EC at 5% O2, scale segment of 100 microns; (b) - viability of EC Representative histograms are presented (the lower left quadrant is a population of living cells, the upper left quadrant is cells in a state of early apoptosis, the upper right quadrant is cells in a state of late apoptosis, the lower right quadrant is cells in a state of necrosis); (c) – expression of adhesion molecules on EC at different O2 levels, representative histograms at 5% O2. ECs – intact EC, ECs TNF-α – activated EC, control cells stained with isotypic IgG1 antibodies.

Download (381KB)
5. Fig. 4. Morphology and transcription profile of MSCs after interaction with EC activated TNF-α. (a) – type of co-culture after 24 hours at 5 and 0.1% O2. Representative images obtained by combining microphotographs taken in phase contrast and fluorescence mode are presented, endothelial cells are stained with fluorescent dye PKH27. The scale segment is 100 microns. The arrows indicate MSC; (b), (c) – expression of MSC differentiation genes after interaction with EC activated at a reduced O2 content. The graphs show the multiplicity of changes in gene expression in MSCs after co-cultivation compared with the monoculture of MSCs (dotted line) under the same conditions. The data are presented as median, interquartile range, maximum and minimum values, n ≥ 3. * – p < 0.05 compared with the monoculture of MSC cultivated at 5% O2. MSC+EC TNF-α –MSC after 24 hours of co-cultivation with activated TNF-α EC.

Download (356KB)
6. Fig. 5. Expression of MSCs intercellular interaction molecules after co-cultivation with TNF-α activated EC. The graphs show the proportion of positively colored MSCs before and after interaction with activated EC (median, minimum and maximum values). In the same graphs, the size of the bubbles corresponds to the value of the median fluorescence intensity of the stained cells, n ≥ 3. * – p < 0.05 compared with the monoculture of MSC under the same cultivation conditions. 1 – MSK 5% O2, 2 – MSK after co–cultivation with EC 5% O2, 3 – MSK 0.1% O2, 4 - MSK after co-cultivation with EC 0.1% O2.

Download (158KB)
7. Fig. 6. Changes in the paracrine activity of EC and MSC after interaction with a reduced O2 content. (a) – concentration of IL-6 in the medium from mono- and co–cultures of MSC and activated EC; (b) - concentration of IL-8 in the medium from mono- and co-cultures of MSC and activated EC; (c) – changes in the expression of IL6 and IL8 genes in MSCs; (d) – a change in the expression of IL6 and IL8 genes in the EC. The graphs show the multiplicity of differences in the expression of genes of interest in MSCs (c) and EC (d) after interaction compared with a monoculture of cells (dotted line) under the same cultivation conditions. The data are presented as median, interquartile range, maximum and minimum values, n = 3, * – p < 0.05 compared with the MSC monoculture under the same cultivation conditions. ** – p < 0.05 compared to the EC monoculture under the same cultivation conditions. MSC+EC TNF-α – MSC after interaction with activated EC. EC TNF-α+MSC – activated ECS after interaction with MSC.

Download (221KB)
8. Fig. 7. Paracrine effect of EC on the mobility of MSCs. (a) – non-directional migration of MSCs in the "wound" model in an air-conditioned environment from activated ECS. Representative micrographs immediately after the application of the "wound" (0 h) and a day later (24 h); (b) – the area of the wound closure. On the graph, the closing area is represented in % relative to the original area of the wound; (c) – directed migration of MSCs through the transwell membrane in an air-conditioned environment from activated ECS. Representative micrographs taken after 24 hours of incubation of cells in the wells of tablets; (d) – the graph shows the proportion of cells that migrated in 24 hours (the ratio of cells that migrated in the conditioned environment from EC, compared with the number of cells that migrated in the growth medium). The data are presented as median, interquartile range, maximum and minimum values, n = 3. * – p < 0.05 compared with the number of cells migrated in the growth medium (Cntrl). CM 5% O2 – conditioned environment from activated EC,

Download (225KB)
9. Fig. 8. Determination of the level of proteases in MSC lysates before and after 24 hours of co-cultivation with EC activated TNF-α. (a) – representative photographs of membranes; (b), (c) – the layout and names of detecting antibodies to various proteases.

Download (203KB)
10. Fig. 9. Effects of hypoxic stress on the interaction of activated EC and MSCs.

Download (199KB)

Copyright (c) 2024 Russian Academy of Sciences

This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies