Expression profile of the isogenic early mesodermal cells differentiated from induced pluripotent human stem cells

Cover Page

Cite item

Full Text

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

Abstract

Scar formation during normal regeneration of damaged tissue can lead to noticeable cosmetic and functional defects of organs and significantly affect the quality of life. However, it is known that fetal tissues before the third trimester of pregnancy are capable of complete regeneration with the restoration of the original architecture and functional activity. Understanding the cellular and molecular mechanisms of fetal wound regeneration will provide the basis for the development of successful treatments aimed to minimize scarring. Mesenchymal stromal cells (MSCs) play an important role in tissue repair, since the cytokines, chemokines, growth factors and extracellular vesicles they secrete are involved in the regulation of migration, angiogenesis, synthesis and remodeling of the extracellular matrix. Mesodermal differentiation of human induced pluripotent stem cells (iPSCs) makes possible to reproduce the successive stages of embryogenesis in vitro and to create isogenic cell models of MSCs corresponding to different stages of human development. In this work, we performed the directed multistage mesodermal differentiation of iPSCs into isogenic cell lines of the primitive streak, lateral and paraxial mesoderm and a comparative analysis of their expression profiles was carried out. It was shown that the resulting cells of the lateral mesoderm (LM) and paraxial mesoderm (PM) are precursors for MSCs. MSCs obtained as a result of differentiation of both LM and PM cells had a similar profile for the expression of pan-mesodermal markers. Comparative analysis of the functional activity of MSCs and their precursors in a pro-inflammatory microenvironment will provide molecular tools for a better understanding of the fundamental mechanisms of fetal tissue regeneration and identify therapeutic targets to minimize scarring and pathological processes characterized by excessive fibroplasia.

Full Text

Restricted Access

About the authors

А. V. Selezneva

Vavilov Institute of General Genetics of the Russian Academy of Sciences

Email: yu_suzdaltseva@mail.ru
Russian Federation, Moscow

Е. V. Korobko

Vavilov Institute of General Genetics of the Russian Academy of Sciences

Email: yu_suzdaltseva@mail.ru
Russian Federation, Moscow

S. L. Kiselev

Vavilov Institute of General Genetics of the Russian Academy of Sciences

Email: yu_suzdaltseva@mail.ru
Russian Federation, Moscow

Y. G. Suzdaltseva

Vavilov Institute of General Genetics of the Russian Academy of Sciences

Author for correspondence.
Email: yu_suzdaltseva@mail.ru
Russian Federation, Moscow

References

  1. Shaw TJ, Martin P (2009) Wound repair at a glance. J Cell Sci 122(Pt 18):3209–3213. https://doi.org/10.1242/jcs.031187
  2. Marshall CD, Hu MS, Leavitt T, Barnes LA, Lorenz HP, Longaker MT (2018). Cutaneous Scarring: Basic Science, Current Treatments, and Future Directions. Adv Wound Care (New Rochelle) 7(2):29–45. https://doi.org/10.1089/wound.2016.0696
  3. Hu MS, Maan ZN, Wu JC, Rennert RC, Hong WX, Lai TS, Cheung AT, Walmsley GG, Chung MT, McArdle A, Longaker MT, Lorenz HP (2014) Tissue engineering and regenerative repair in wound healing. Ann Biomed Eng 42(7):1494–1507. https://doi.org/10.1007/s10439-014-1010-z
  4. Kim EY, Hussain A, Khachemoune A (2022) Evidence-based management of keloids and hypertrophic scars in dermatology. Arch Dermatol Res 315(6):1487–1495. https://doi.org/10.1007/s00403-022-02509-x
  5. Colwell AS, Longaker MT, Lorenz HP (2003) Fetal wound healing. Front Biosci 1;8: s1240–1248. https://doi.org/10.2741/1183
  6. Moore AL, Marshall CD, Barnes LA, Murphy MP, Ransom RC, Longaker MT (2018) Scarless wound healing: Transitioning from fetal research to regenerative healing. Wiley Interdiscip Rev Dev Biol 7(2):10.1002/wdev.309. https://doi.org/10.1002/wdev.309
  7. Suzdaltseva Y, Kiselev SL (2023) Mesodermal Derivatives of Pluripotent Stem Cells Route to Scarless Healing. Int J Mol Sci 24(15):11945. https://doi.org/10.3390/ijms241511945
  8. Pittenger MF, Discher DE, Péault BM, Phinney DG, Hare JM, Caplan AI (2019) Mesenchymal stem cell perspective: cell biology to clinical progress. NPJ Regen Med 4:22. https://doi.org/10.1038/s41536-019-0083-6. eCollection 2019
  9. Fu Y, Karbaat L, Wu L, Leijten J, Both SK, Karperien M (2017) Trophic Effects of Mesenchymal Stem Cells in Tissue Regeneration. Tissue Eng Part B Rev 23(6):515–528. https://doi.org/10.1089/ten.TEB.2016.0365
  10. Han Y, Yang J, Fang J, Zhou Y, Candi E, Wang J, Hua D, Shao C, Shi Y (2022) The secretion profile of mesenchymal stem cells and potential applications in treating human diseases. Signal Transduct Target Ther 7(1):92. https://doi.org/10.1038/s41392-022-00932-0
  11. Kusuma GD, Carthew J, Lim R, Frith JE (2017) Effect of the Microenvironment on Mesenchymal Stem Cell Paracrine Signaling: Opportunities to Engineer the Therapeutic Effect. Stem Cells Dev 26(9):617–631. https://doi.org/10.1089/scd.2016.0349
  12. Le Blanc K, Davies LC (2015) Mesenchymal stromal cells and the innate immune response. Immunol Lett 168(2):140–146. https://doi.org/10.1016/j.imlet.2015.05.004
  13. Weiss ARR, Dahlke MH (2019) Immunomodulation by Mesenchymal Stem Cells (MSCs): Mechanisms of Action of Living, Apoptotic, and Dead MSCs. Front Immunol 10:1191. https://doi.org/10.3389/fimmu.2019.01191
  14. Suzdaltseva Y, Goryunov K, Silina E, Manturova N, Stupin V, Kiselev SL (2022) Equilibrium among Inflammatory Factors Determines Human MSC-Mediated Immunosuppressive Effect. Cells 11(7):1210. https://doi.org/10.3390/cells11071210
  15. Guillén MI, Platas J, Pérez Del Caz MD, Mirabet V, Alcaraz MJ (2018) Paracrine Anti-inflammatory Effects of Adipose Tissue-Derived Mesenchymal Stem Cells in Human Monocytes. Front Physiol 31(9):661. https://doi.org/10.3389/fphys.2018.00661
  16. Mareschi K, Castiglia S, Sanavio F, Rustichelli D, Muraro M, Defedele D, Bergallo M, Fagioli F (2016) Immunoregulatory effects on T lymphocytes by human mesenchymal stromal cells isolated from bone marrow, amniotic fluid, and placenta. Exp Hematol 44(2):138–150.e1. https://doi.org/10.1016/j.exphem.2015.10.009
  17. Jiang D, Scharffetter-Kochanek K (2020) Mesenchymal Stem Cells Adaptively Respond to Environmental Cues Thereby Improving Granulation Tissue Formation and Wound Healing. Front Cell Dev Biol 8:697. https://doi.org/10.3389/fcell.2020.00697
  18. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop Dj, Horwitz E (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8(4):315–317. https://doi.org/10.1080/14653240600855905
  19. Cheung C, Bernardo AS, Trotter MW, Pedersen RA, Sinha S (2012) Generation of human vascular smooth muscle subtypes provides insight into embryological origin-dependent disease susceptibility. Nat Biotechnol 30(2):165–173. https://doi.org/10.1038/nbt.2107
  20. Isern J, García-García A, Martín AM, Arranz L, Martín-Pérez D, Torroja C, Sánchez-Cabo F, Méndez-Ferrer S (2014) The neural crest is a source of mesenchymal stem cells with specialized hematopoietic stem cell niche function. Elife 3: e03696. https://doi.org/10.7554/eLife.03696
  21. Sheng G (2015) The developmental basis of mesenchymal stem/stromal cells (MSCs). BMC Dev Biol 15:44. https://doi.org/10.1186/s12861–015–0094–5
  22. Shutova MV, Bogomazova AN, Lagarkova MA, Kiselev SL (2009) Generation and characterization of human induced pluripotent stem cells. Acta Naturae 1(2):91–92. https:// PMC3347519
  23. Loh KM, Chen A, Koh PW, Deng TZ, Sinha R, Tsai JM, Barkal AA, Shen KY, Jain R, Morganti RM, Shyh-Chang N, Fernhoff NB, George BM, Wernig G, Salomon REA, Chen Z, Vogel H, Epstein JA, Kundaje A, Talbot WS, Beachy PA, Ang LT, Weissman IL (2016) Mapping the Pairwise Choices Leading from Pluripotency to Human Bone, Heart, and Other Mesoderm Cell Types. Cell 166(2):451–467. https://doi.org/10.1016/j.cell.2016.06.011
  24. Tran NT, Trinh QM, Lee GM, Han YM (2012) Efficient differentiation of human pluripotent stem cells into mesenchymal stem cells by modulating intracellular signaling pathways in a feeder/serum-free system. Stem Cells Dev 21(7):1165–1175. https://doi.org/10.1089/scd.2011.0346
  25. Xi H, Fujiwara W, Gonzalez K, Jan M, Liebscher S, Van Handel B, Schenke-Layland K, Pyle AD (2017) In Vivo Human Somitogenesis Guides Somite Development from hPSCs. Cell Rep 18(6):1573–1585. https://doi.org/10.1016/j.celrep.2017.01.040
  26. Nakajima T, Shibata M, Nishio M, Nagata S, Alev C, Sakurai H, Toguchida J, Ikeya M (2018) Modeling human somite development and fibrodysplasia ossificans progressiva with induced pluripotent stem cells. Development 145(16): dev165431. https://doi.org/10.1242/dev.165431
  27. Burrington JD (1971) Wound healing in the fetal lamb. J Pediatr Surg 6(5):523–528. https://doi.org/10.1016/0022-3468(71)90373-3
  28. Somasundaram K, Prathap K (1970) Intra-uterine healing of skin wounds in rabbit foetuses. J Pathol 100(2):81–86. https://doi.org/10.1002/path.1711000202
  29. Goss AN (1977) Intra-uterine healing of fetal rat oral mucosal, skin and cartilage wounds. J Oral Pathol 6(1):35–43. https://doi.org/10.1111/j.1600-0714.1977.tb01792.x
  30. Gnyawali SC, Sinha M, El Masry MS, Wulff B, Ghatak S, Soto-Gonzalez F, Wilgus TA, Roy S, Sen CK (2020) High resolution ultrasound imaging for repeated measure of wound tissue morphometry, biomechanics and hemodynamics under fetal, adult and diabetic conditions. PLoS One 15(11): e0241831. https://doi.org/10.1371/journal.pone.0241831
  31. Lorenz HP, Longaker MT, Perkocha LA, Jennings RW, Harrison MR, Adzick NS (1992) Scarless wound repair: a human fetal skin model. Development 114(1):253–259. https://doi.org/10.1242/dev.114.1.253
  32. Estes JM, Vande Berg JS, Adzick NS, MacGillivray TE, Desmoulière A, Gabbiani G (1994) Phenotypic and functional features of myofibroblasts in sheep fetal wounds. Differentiation 56(3):173–181. https://doi.org/10.1046/j.1432-0436.1994.5630173.x
  33. Cass DL, Sylvester KG, Yang EY, Crombleholme TM, Adzick NS (1997) Myofibroblast persistence in fetal sheep wounds is associated with scar formation. J Pediatr Surg 32(7):1017–1021. https://doi.org/10.1016/s0022-3468(97)90390-0
  34. Satish L, Johnson S, Wang JH, Post JC, Ehrlich GD, Kathju S (2010) Chaperonin containing T-complex polypeptide subunit eta (CCT-eta) is a specific regulator of fibroblast motility and contractility. PLoS One 5(4): e10063. https://doi.org/10.1371/journal.pone.0010063
  35. Moulin V, Tam BY, Castilloux G, Auger FA, O'Connor-McCourt MD, Philip A, Germain L (2001) Fetal and adult human skin fibroblasts display intrinsic differences in contractile capacity. J Cell Physiol 188(2):211–222. https://doi.org/10.1002/jcp.1110
  36. Jerrell RJ, Leih MJ, Parekh A (2019) The altered mechanical phenotype of fetal fibroblasts hinders myofibroblast differentiation. Wound Repair Regen 27(1):29–38. https://doi.org/ 10.1111/wrr.12677
  37. Brink HE, Miller GJ, Beredjiklian PK, Nicoll SB (2006) Serum-dependent effects on adult and fetal tendon fibroblast migration and collagen expression. Wound Repair Regen 14(2):179–186. https://doi.org/10.1111/j.1743-6109.2006.00108.x
  38. Nekrasov ED, Vigont VA, Klyushnikov SA, Lebedeva OS, Vassina EM, Bogomazova AN, Chestkov IV, Semashko TA, Kiseleva E, Suldina LA, Bobrovsky PA, Zimina OA, Ryazantseva MA, Skopin AY, Illarioshkin SN, Kaznacheyeva EV, Lagarkova MA, Kiselev SL (2016) Manifestation of Huntington's disease pathology in human induced pluripotent stem cell-derived neurons. Mol Neurodegener 11:27. https://doi.org/10.1186/s13024-016-0092-5
  39. Philonenko ES, Shutova MV, Khomyakova EA, Vassina EM, Lebedeva OS, Kiselev SL, Lagarkova MA (2017) Differentiation of Human Pluripotent Stem Cells into Mesodermal and Ectodermal Derivatives Is Independent of the Type of Isogenic Reprogrammed Somatic Cells. Acta Naturae 9(1):68–74. https://PMC5406662
  40. Panova AV, Klementieva NV, Sycheva AV, Korobko EV, Sosnovtseva AO, Krasnova TS, Karpova MR, Rubtsov PM, Tikhonovich YV, Tiulpakov AN, Kiselev SL (2022) Aberrant Splicing of INS Impairs Beta-Cell Differentiation and Proliferation by ER Stress in the Isogenic iPSC Model of Neonatal Diabetes. Int J Mol Sci 23(15):8824. https://doi.org/10.3390/ijms23158824
  41. Chijimatsu R, Ikeya M, Yasui Y, Ikeda Y, Ebina K, Moriguchi Y, Shimomura K, Hart DA, Hideki Y, Norimasa N (2017) Characterization of Mesenchymal Stem Cell-Like Cells Derived From Human iPSCs via Neural Crest Development and Their Application for Osteochondral Repair. Stem Cells Int 2017:1960965. https://doi.org/10.1155/2017/1960965
  42. Kimura M, Furukawa H, Shoji M, Shinozawa T (2019) Increased mesodermal and mesendodermal populations by BMP4 treatment facilitates human iPSC line differentiation into a cardiac lineage. J Stem Cells Regen Med 15(2):45–51. https://doi.org/10.46582/jsrm.1502009
  43. Wang Y, Wang H, Guo J, Gao J, Wang M, Xia M, Wen Y, Su P, Yang M, Liu M, Shi L, Cheng T, Zhou W, Zhou J (2020) LGR4, Not LGR5, Enhances hPSC Hematopoiesis by Facilitating Mesoderm Induction via TGF-Beta Signaling Activation. Cell Rep 31(5):107600. https://doi.org/10.1016/j.celrep.2020.107600
  44. Kamatani T, Hagizawa H, Yarimitsu S, Morioka M, Koyamatsu S, Sugimoto M, Kodama J, Yamane J, Ishiguro H, Shichino S, Abe K, Fujibuchi W, Fujie H, Kaito T, Tsumaki N (2022) Human iPS cell-derived cartilaginous tissue spatially and functionally replaces nucleus pulposus. Biomaterials 284:121491. https://doi.org/10.1016/j.biomaterials.2022.121491
  45. Philonenko ES, Tan Y, Wang C, Zhang B, Shah Z, Zhang J, Ullah H, Kiselev SL, Lagarkova MA, Li D, Dai Y, Samokhvalov IM (2021) Recapitulative haematopoietic development of human pluripotent stem cells in the absence of exogenous haematopoietic cytokines. J Cell Mol Med 25(18):8701–8714. https://doi.org/10.1111/jcmm.16826
  46. Nakajima T, Ikeya M (2021). Development of pluripotent stem cell-based human tenocytes. Dev Growth Differ 63(1):38–46. https://doi.org/10.1111/dgd.12702
  47. Liu TM, Yildirim ED, Li P, Fang HT, Denslin V, Kumar V, Loh YH, Lee EH, Cool SM, Teh BT, Hui JH, Lim B, Shyh-Chang N (2020) Ascorbate and Iron Are Required for the Specification and Long-Term Self-Renewal of Human Skeletal Mesenchymal Stromal Cells. Stem Cell Reports 14(2):210–225. https://doi.org/10.1016/j.stemcr.2020.01.002
  48. Fukuta M, Nakai Y, Kirino K, Nakagawa M, Sekiguchi K, Nagata S, Matsumoto Y, Yamamoto T, Umeda K, Heike T, Okumura N, Koizumi N, Sato T, Nakahata T, Saito M, Otsuka T, Kinoshita S, Ueno M, Ikeya M, Toguchida J (2014) Derivation of mesenchymal stromal cells from pluripotent stem cells through a neural crest lineage using small molecule compounds with defined media. PLoS One 9(12): e112291. https://doi.org/10.1371/journal.pone.0112291. eCollection 2014
  49. Wang H, Li D, Zhai Z, Zhang X, Huang W, Chen X, Huang L, Liu H, Sun J, Zou Z, Fan Y, Ke Q, Lai X, Wang T, Li X, Shen H, Xiang AP, Li W (2019) Characterization and Therapeutic Application of Mesenchymal Stem Cells with Neuromesodermal Origin from Human Pluripotent Stem Cells. Theranostics 9(6):1683–1697. https://doi.org/10.7150/thno.30487. eCollection 2019
  50. Wei Y, Wang B, Jia L, Huang W, Xiang AP, Fang C, Liang X, Li W (2022) Lateral Mesoderm-Derived Mesenchymal Stem Cells With Robust Osteochondrogenic Potential and Hematopoiesis-Supporting Ability. Front Mol Biosci 9:767536. https://doi.org/10.3389/fmolb.2022.767536. eCollection 2022
  51. Umeda K, Zhao J, Simmons P, Stanley E, Elefanty A, Nakayama N (2012) Human chondrogenic paraxial mesoderm, directed specification and prospective isolation from pluripotent stem cells. Sci Rep 2:455. https://doi.org/10.1038/srep00455
  52. Kishimoto K, Iwasawa K, Sorel A, Ferran-Heredia C, Han L, Morimoto M, Wells JM, Takebe T, Zorn AM (2022) Directed differentiation of human pluripotent stem cells into diverse organ-specific mesenchyme of the digestive and respiratory systems. Nat Protoc 17(11):2699–2719. https://doi.org/10.1038/s41596-022-00733-3
  53. Smith CA, Humphreys PA, Naven MA, Woods S, Mancini FE, O'Flaherty J, Meng QJ, Kimber SJ (2023) Directed differentiation of hPSCs through a simplified lateral plate mesoderm protocol for generation of articular cartilage progenitors. PLoS One 18(1): e0280024. https://doi.org/10.1371/journal.pone.0280024
  54. Kimbrel EA, Kouris NA, Yavanian GJ, Chu J, Qin Y, Chan A, Singh RP, McCurdy D, Gordon L, Levinson RD, Lanza R (2014) Mesenchymal stem cell population derived from human pluripotent stem cells displays potent immunomodulatory and therapeutic properties. Stem Cells Dev 23(14):1611–1624. https://doi.org/10.1089/scd.2013.0554
  55. Eto S, Goto M, Soga M, Kaneko Y, Uehara Y, Mizuta H, Era T (2018) Mesenchymal stem cells derived from human iPS cells via mesoderm and neuroepithelium have different features and therapeutic potentials. PLoS One 13(7): e0200790. https://doi.org/10.1371/journal.pone.0200790. eCollection 2018
  56. Spitzhorn LS, Megges M, Wruck W, Rahman MS, Otte J, Degistirici Ö, Meisel R, Sorg RV, Oreffo ROC, Adjaye J (2019) Human iPSC-derived MSCs (iMSCs) from aged individuals acquire a rejuvenation signature. Stem Cell Res Ther 10(1):100. https://doi.org/10.1186/s13287-019-1209-x
  57. Wruck W, Graffmann N, Spitzhorn LS, Adjaye J (2021) Human Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cells Acquire Rejuvenation and Reduced Heterogeneity. Front Cell Dev Biol 9:717772. https://doi.org/10.3389/fcell.2021.717772
  58. Billing AM, Ben Hamidane H, Dib SS, Cotton RJ, Bhagwat AM, Kumar P, Hayat S, Yousri NA, Goswami N, Suhre K, Rafii A, Graumann J (2016) Comprehensive transcriptomic and proteomic characterization of human mesenchymal stem cells reveals source specific cellular markers. Sci Rep 6:21507. https://doi.org/10.1038/srep21507
  59. Liu Y, Goldberg AJ, Dennis JE, Gronowicz GA, Kuhn LT (2012) One-step derivation of mesenchymal stem cell (MSC)-like cells from human pluripotent stem cells on a fibrillar collagen coating. PLoS One 7(3): e33225. https://doi.org/10.1371/journal.pone.0033225
  60. Villa-Diaz LG, Brown SE, Liu Y, Ross AM, Lahann J, Parent JM, Krebsbach PH (2012) Derivation of mesenchymal stem cells from human induced pluripotent stem cells cultured on synthetic substrates. Stem Cells 30(6):1174–1181. https://doi.org/10.1002/stem.1084
  61. Diederichs S, Tuan RS (2014) Functional comparison of human-induced pluripotent stem cell-derived mesenchymal cells and bone marrow-derived mesenchymal stromal cells from the same donor. Stem Cells Dev 23(14):1594–1610. https://doi.org/10.1089/scd.2013.0477
  62. Rubtsov Y, Goryunov К, Romanov А, Suzdaltseva Y, Sharonov G, Tkachuk V (2017) Molecular Mechanisms of Immunomodulation Properties of Mesenchymal Stromal Cells: A New Insight into the Role of ICAM-1. Stem Cells Int 2017:6516854. https://doi.org/10.1155/2017/6516854.
  63. Suzdaltseva YG, Goryunov KV, Rubtsov YP (2018) The Role of Intercellular Contacts in Induction of Indolamine-2.3-Dioxygenase Synthesis in MMSC from Adipose Tissue. Cell and Tissue Biology 12: 391–401. https://doi.org/10.1134/S1990519X18050085
  64. Suzdaltseva Y, Zhidkih S, Kiselev SL, Stupin V (2020) Locally Delivered Umbilical Cord Mesenchymal Stromal Cells Reduce Chronic Inflammation in Long-Term Nonhealing Wounds: A Randomized Study. Stem Cells Int 2020:5308609. https://doi.org/10.1155/2020/5308609.

Supplementary files

Supplementary Files
Action
1. JATS XML
2. Fig. 1. Morphological changes in cells during differentiation of iPSCs in the mesodermal direction. (a) – initial iPSC culture. (b) – morphological changes in cells during induction of iPSC differentiation into paraxial mesoderm. (c) – MSCs obtained from paraxial mesoderm cells. (d) – morphological changes in cells during induction of iPSC differentiation into lateral mesoderm. (e) – MSCs obtained from lateral mesoderm cells. Phase contrast. iPSC – induced pluripotent stem cells. MSC – mesenchymal stromal cells.

Download (554KB)
3. Fig. 2. Expression of pluripotency markers during differentiation of iPSCs in the mesodermal direction. (a) – Expression of the POU5F1, SOX2, NANOG genes in the initial iPSC culture. Electrophoregram of PCR products. Changes in the expression of pluripotency markers in iPSCs during differentiation into paraxial mesoderm (b) and lateral mesoderm (c). The average values ​​and their errors of 3 independent experiments are shown. PM – paraxial mesoderm. LM – lateral mesoderm.

Download (103KB)
4. Fig. 3. Expression of primitive streak genes in lateral and paraxial mesoderm cells differentiated from iPSCs. (a) – co-expression of TBXT and MIXL1 genes in LM and PM cells on day 2 of cultivation. Electrophoregram of PCR products. (b) and (c) – dynamics of changes in TBXT and MIXL1 gene expression during differentiation of iPSCs into PM and LM. Average values ​​and their errors of 3 independent experiments are shown. PM – paraxial mesoderm. LM – lateral mesoderm.

Download (86KB)
5. Fig. 4. Expression of the HAND1, HAND2, BMP4 genes in LM cells differentiated from iPSCs. (a) – the level of gene expression in iPSCs, LM and PM cells on the 7th day of cultivation. Electropherogram of PCR products. Changes in the expression of the HAND1 (b), HAND2 (c), BMP4(d) genes during differentiation of iPSCs into LM. The mean values ​​and their errors of 3 independent experiments are shown. The asterisk indicates the significance of differences at p < 0.05. iPSC – induced pluripotent stem cells. PM – paraxial mesoderm. LM – lateral mesoderm.

Download (125KB)
6. Fig. 5. Relative gene expression in PM and LM cells differentiated from iPSCs. The expression level of the HAND1 (a), HAND2 (b), BMP4 (c), and WNT5A (d) genes on the 7th day of differentiation relative to the initial iPSC culture. The mean values ​​and their errors of 3 independent experiments are shown. The asterisk indicates the significance of differences at p < 0.05. (e) – expression of the presomitic mesoderm gene MEOX1 in cells during specific multistage and nonspecific single-stage differentiation of iPSCs in the mesodermal direction on the 7th day of cultivation. iPSC – induced pluripotent stem cells. PM – paraxial mesoderm. LM – lateral mesoderm. 1 – LM cells, 2 – PM cells, 3 – cells obtained as a result of spontaneous mesodermal differentiation of iPSCs in a standard medium for culturing MSCs.

Download (114KB)

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