Skip to main content

Advertisement

Log in

Regulatory role of apelin receptor signaling in migration and differentiation of mouse embryonic stem cell-derived mesoderm cells and mesenchymal stem/stromal cells

  • Research Article
  • Published:
Human Cell Aims and scope Submit manuscript

Abstract

Mesoderm-derived cells, including bone, muscle, and mesenchymal stem/stromal cells (MSCs), constitute various parts of vertebrate body. Cell therapy with mesoderm specification in vitro may be a promising treatment for diseases affecting organs of mesodermal origin. Repair and regeneration of damaged organs with in vitro generation of mesoderm-derived tissues and MSCs hold a great potential for regenerative therapy. Therefore, understanding the signaling pathways involving mesoderm and mesoderm-derived cellular differentiation is important. Previous findings indicated the importance of Apelin receptor (Aplnr) signaling, during embryonic development, in gastrulation, cell migration, and differentiation. Nevertheless, regulatory role of Aplnr pathway in differentiation of mesoderm and mesoderm-derived MSCs remains unclear. In the current study, we tried to elucidate the role of Aplnr signaling during mesoderm cell migration and differentiation from mouse embryonic stem cells (mESCs). By activating and suppressing Aplnr signaling pathway via peptide, small molecule, and genetic modifications including siRNA- and shRNA-mediated knockdown and CRISPR-Cas9-mediated knockout (KO), we revealed that Aplnr signaling not only induces migration of cells during germ layer formation but also enhances mesoderm differentiation through FGF/MAPK pathway. Antibody array and LC/MS protein profiling data demonstrated that Apelin-13 treatment enhanced cell cycle, EGFR, FGF, Wnt, and Integrin signaling pathway proteins. Furthermore, Aplelin-13 treatment improved MSC characteristics, with mesenchymal phenotype and high expression of MSC markers, and silencing Aplnr signaling components resulted in significantly reduced expression of MSC markers. Also, Aplnr signaling activity enhanced proliferation and survival of the cells during MSC derivation from mesoderm.

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
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

Data availability

The datasets generated during and/or analyzed during the current study are not publicly available due to [Because all data were represented in the article. There are no datasets which need to be publicly shared. Raw data can be requested from corresponding author], but are available from the corresponding author on reasonable request.

References

  1. Murry CE, Keller G. Differentiation of embryonic stem cells to clinically relevant populations: lessons from embryonic development. Cell. 2008;132(4):661–80.

    Article  CAS  PubMed  Google Scholar 

  2. Pouton CW, Haynes JM. Embryonic stem cells as a source of models for drug discovery. Nat Rev Drug Discov. 2007;6(8):605–16.

    Article  CAS  PubMed  Google Scholar 

  3. Bratt-Leal AM, Carpenedo RL, McDevitt TC. Engineering the embryoid body microenvironment to direct embryonic stem cell differentiation. Biotechnol Prog. 2009;25(1):43–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Itskovitz-Eldor J, Schuldiner M, Karsenti D, Eden A, Yanuka O, Amit M, et al. Differentiation of human embryonic stem cells into embryoid bodies comprising the three embryonic germ layers. Mol Med. 2000;6(2):88–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Locascio A, Nieto MA. Cell movements during vertebrate development: integrated tissue behaviour versus individual cell migration. Curr Opin Genet Dev. 2001;11(4):464–9.

    Article  CAS  PubMed  Google Scholar 

  6. Ferrer-Vaquer A, Viotti M, Hadjantonakis A-K. Transitions between epithelial and mesenchymal states and the morphogenesis of the early mouse embryo. Cell Adh Mig. 2010;4(3):447–57.

    Article  Google Scholar 

  7. Arnold SJ, Robertson EJ. Making a commitment: cell lineage allocation and axis patterning in the early mouse embryo. Nat Rev Mol Cell Biol. 2009;10(2):91–103.

    Article  CAS  PubMed  Google Scholar 

  8. Ferrer-Vaquer A, Hadjantonakis AK. Birth defects associated with perturbations in preimplantation, gastrulation, and axis extension: from conjoined twinning to caudal dysgenesis. Wiley Interdiscip Rev Dev Biol. 2013;2(4):427–42.

    Article  PubMed  Google Scholar 

  9. Li C, Zhao H, Wang B. Mesenchymal stem/stromal cells: developmental origin, tumorigenesis and translational cancer therapeutics. Transl Oncol. 2021;14(1):100948.

    Article  CAS  PubMed  Google Scholar 

  10. Vodyanik MA, Yu J, Zhang X, Tian S, Stewart R, Thomson JA, et al. A mesoderm-derived precursor for mesenchymal stem and endothelial cells. Cell Stem Cell. 2010;7(6):718–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wei Y, Wang B, Jia L, Huang W, Xiang AP, Fang C, et al. Lateral Mesoderm-derived mesenchymal stem cells with robust osteochondrogenic potential and hematopoiesis-supporting ability. Front Mol Biosci. 2022;9:767536

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Liu T-M. Application of mesenchymal stem cells derived from human pluripotent stem cells in regenerative medicine. World J Stem Cells. 2021;13(12):1826.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Witman N, Sahara M. Cardiac progenitor cells in basic biology and regenerative medicine. Stem Cells Int. 2018;2018:8283648.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Tani S, Chung U-I, Ohba S, Hojo H. Understanding paraxial mesoderm development and sclerotome specification for skeletal repair. Exp Mol Med. 2020;52(8):1166–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Araoka T, Mae S-I, Kurose Y, Uesugi M, Ohta A, Yamanaka S, et al. Efficient and rapid induction of human iPSCs/ESCs into nephrogenic intermediate mesoderm using small molecule-based differentiation methods. PloS One. 2014;9(1):e84881.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Grunz H. The vertebrate organizer. Springer Sci Bus Media; 2004.

    Book  Google Scholar 

  17. Zhang X, Chen Y, Ye Y, Wang J, Wang H, Yuan G, et al. Wnt signaling promotes hindgut fate commitment through regulating multi-lineage genes during hESC differentiation. Cell Signal. 2017;29:12–22.

    Article  CAS  PubMed  Google Scholar 

  18. O’Dowd BF, Heiber M, Chan A, Heng HH, Tsui L-C, Kennedy JL, et al. A human gene that shows identity with the gene encoding the angiotensin receptor is located on chromosome 11. Gene. 1993;136(1–2):355–60.

    Article  CAS  PubMed  Google Scholar 

  19. Tatemoto K, Hosoya M, Habata Y, Fujii R, Kakegawa T, Zou M-X, et al. Isolation and characterization of a novel endogenous peptide ligand for the human APJ receptor. Biochem Biophys Res Commun. 1998;251(2):471–6.

    Article  CAS  PubMed  Google Scholar 

  20. Pauli A, Norris ML, Valen E, Chew G-L, Gagnon JA, Zimmerman S, et al. Toddler: an embryonic signal that promotes cell movement via Apelin receptors. Science. 2014;343(6172):1248636.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Şişli HB, Hayal TB, Şenkal S, Kıratlı B, Sağraç D, Seçkin S, et al. Apelin receptor signaling protects GT1-7 GnRH neurons against oxidative stress in vitro. Cell Mol Neurobiol. 2020;42:1–23.

    Google Scholar 

  22. Mesmin C, Fenaille F, Becher F, Tabet J-C, Ezan E. Identification and characterization of apelin peptides in bovine colostrum and milk by liquid chromatography–mass spectrometry. J Proteome Res. 2011;10(11):5222–31.

    Article  CAS  PubMed  Google Scholar 

  23. Shin K, Pandey A, Liu X-Q, Anini Y, Rainey JK. Preferential apelin-13 production by the proprotein convertase PCSK3 is implicated in obesity. FEBS Open Bio. 2013;3:328–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Kawamata Y, Habata Y, Fukusumi S, Hosoya M, Fujii R, Hinuma S, et al. Molecular properties of apelin: tissue distribution and receptor binding. Biochim Biophys Acta Mol Cell Res. 2001;1538(2–3):162–71.

    Article  CAS  Google Scholar 

  25. Liu W, Yan J, Pan W, Tang M. Apelin/Elabela-APJ: a novel therapeutic target in the cardiovascular system. Ann Transl Med. 2020;8(5):243.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Chng SC, Ho L, Tian J, Reversade B. ELABELA: a hormone essential for heart development signals via the apelin receptor. Dev Cell. 2013;27(6):672–80.

    Article  CAS  PubMed  Google Scholar 

  27. Zeng X-XI, Wilm TP, Sepich DS, Solnica-Krezel L. Apelin and its receptor control heart field formation during zebrafish gastrulation. Dev Cell. 2007;12(3):391–402.

    Article  CAS  PubMed  Google Scholar 

  28. Janssens P, de Loor H, Decuypere J-P, Vennekens R, Llorens-Cortes C, Mekahli D, et al. On methods for the measurement of the apelin receptor ligand apelin. Sci Rep. 2022;12(1):1–13.

    Article  Google Scholar 

  29. Ho L, Tan SY, Wee S, Wu Y, Tan SJ, Ramakrishna NB, et al. ELABELA is an endogenous growth factor that sustains hESC self-renewal via the PI3K/AKT pathway. Cell Stem Cell. 2015;17(4):435–47.

    Article  CAS  PubMed  Google Scholar 

  30. Norris ML, Pauli A, Gagnon JA, Lord ND, Rogers KW, Mosimann C, et al. Toddler signaling regulates mesodermal cell migration downstream of Nodal signaling. Elife. 2017;6:e22626.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Macrae RG, Colzani MT, Williams TL, Bayraktar S, Kuc RE, Pullinger AL, et al. Inducible apelin receptor knockdown reduces differentiation efficiency and contractility of hESC-derived cardiomyocytes. Cardiovasc Res. 2022. https://doi.org/10.1093/cvr/cvac065.

    Article  PubMed  Google Scholar 

  32. Slukvin II, Vodyanik M. Endothelial origin of mesenchymal stem cells. Cell Cycle. 2011;10(9):1370–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Şişli HB, Şenkal S, Sağraç D, Hayal TB, Doğan A. Feeder-dependent/independent mouse embryonic stem cell culture protocol. Springer; 2021.

    Book  Google Scholar 

  34. Şenkal S, Hayal TB, Sağraç D, Şişli HB, Asutay AB, Kıratlı B, et al. Human ESC-derived neuromesodermal progenitors (NMPs) successfully differentiate into mesenchymal stem cells (MSCs). Stem Cell Rev Rep. 2021;18:278–93.

    Google Scholar 

  35. Sağraç D, Şenkal S, Hayal TB, Şahin F, Çobandede Z, Doğan A. Surface coating materials regulates the attachment and differentiation of mouse embryonic stem cell derived embryoid bodies into mesoderm at culture conditions. Cytotechnology. 2022;74(2):293–307.

    Article  PubMed  Google Scholar 

  36. Sakuma T, Nishikawa A, Kume S, Chayama K, Yamamoto T. Multiplex genome engineering in human cells using all-in-one CRISPR/Cas9 vector system. Sci Rep. 2014;4(1):1–6.

    Article  Google Scholar 

  37. Hayal TB, Dogan A, Sisli HB, Kiratli B, Sahin F. Ubiquitin-specific protease 7 downregulation suppresses breast cancer in vitro. Turkish J Biol. 2020;44(4):145–57.

    Article  CAS  Google Scholar 

  38. Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9(7):671–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Sağraç D, Şenkal S, Hayal TB, Demirci S, Şişli HB, Asutay AB, et al. Protective role of Cytoglobin and Neuroglobin against the Lipopolysaccharide (LPS)-induced inflammation in Leydig cells ex vivo. Reprod Biol. 2022;22(1):100595.

    Article  PubMed  Google Scholar 

  40. Akpinar G, Yoneten KK, Kasap M, Karaoz E. Search for novel plasma membrane proteins as potential biomarkers in human mesenchymal stem cells derived from dental pulp, adipose tissue, bone marrow, and hair follicle. J Membr Biol. 2021;254(4):409–22.

    Article  CAS  PubMed  Google Scholar 

  41. Doğan A, Demirci S, Şahin F. In vitro differentiation of human tooth germ stem cells into endothelial-and epithelial-like cells. Cell Biol Int. 2015;39(1):94–103.

    Article  PubMed  Google Scholar 

  42. Metsalu T, Vilo J. ClustVis: a web tool for visualizing clustering of multivariate data using Principal Component Analysis and heatmap. Nucleic Acids Res. 2015;43(W1):W566–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Tyanova S, Temu T, Sinitcyn P, Carlson A, Hein MY, Geiger T, et al. The Perseus computational platform for comprehensive analysis of (prote) omics data. Nat Methods. 2016;13(9):731–40.

    Article  CAS  PubMed  Google Scholar 

  44. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47(D1):D607–13.

    Article  CAS  PubMed  Google Scholar 

  45. Mi H, Ebert D, Muruganujan A, Mills C, Albou L-P, Mushayamaha T, et al. PANTHER version 16: a revised family classification, tree-based classification tool, enhancer regions and extensive API. Nucleic Acids Res. 2021;49(D1):D394–403.

    Article  CAS  PubMed  Google Scholar 

  46. Thomas PD, Campbell MJ, Kejariwal A, Mi H, Karlak B, Daverman R, et al. PANTHER: a library of protein families and subfamilies indexed by function. Genome Res. 2003;13(9):2129–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Mi H, Muruganujan A, Huang X, Ebert D, Mills C, Guo X, et al. Protocol Update for large-scale genome and gene function analysis with the PANTHER classification system (v. 14.0). Nat Protoc. 2019;14(3):703–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. He Z, Lı JJ, Zhen CH, Feng LY, Dıng XY. Effect of leukemia inhibitory factor on embryonic stem cell differentiation: implications for supporting neuronal differentiation 1. Acta Pharmacol Sin. 2006;27(1):80–90.

    Article  CAS  PubMed  Google Scholar 

  49. Bourillot P-Y, Aksoy I, Schreiber V, Wianny F, Schulz H, Hummel O, et al. Novel STAT3 target genes exert distinct roles in the inhibition of mesoderm and endoderm differentiation in cooperation with Nanog. Stem Cells. 2009;27(8):1760–71.

    Article  CAS  PubMed  Google Scholar 

  50. Deshwar AR, Chng SC, Ho L, Reversade B, Scott IC. The Apelin receptor enhances Nodal/TGFβ signaling to ensure proper cardiac development. Elife. 2016;5:e13758.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Doğan A. Apelin receptor (Aplnr) signaling promotes fibroblast migration. Tissue Cell. 2019;56:98–106.

    Article  PubMed  Google Scholar 

  52. Westphal M, Panza P, Kastenhuber E, Wehrle J, Driever W. Wnt/β-catenin signaling promotes neurogenesis in the diencephalospinal dopaminergic system of embryonic zebrafish. Sci Rep. 2022;12(1):1–19.

    Article  Google Scholar 

  53. Patapoutian A, Reichardt LF. Roles of Wnt proteins in neural development and maintenance. Curr Opin Neurobiol. 2000;10(3):392–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Sakurai H, Okawa Y, Inami Y, Nishio N, Isobe KI. Paraxial mesodermal progenitors derived from mouse embryonic stem cells contribute to muscle regeneration via differentiation into muscle satellite cells. Stem Cells. 2008;26(7):1865–73.

    Article  CAS  PubMed  Google Scholar 

  55. Kishi M, Mizuseki K, Sasai N, Yamazaki H, Shiota K, Nakanishi S, et al. Requirement of Sox2-mediated signaling for differentiation of early Xenopus neuroectoderm. Development. 2000;127(4):791–800.

    Article  CAS  PubMed  Google Scholar 

  56. Roellig D, Tan-Cabugao J, Esaian S, Bronner ME. Dynamic transcriptional signature and cell fate analysis reveals plasticity of individual neural plate border cells. Elife. 2017;6:e21620.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Norrman K, Strömbeck A, Semb H, Ståhlberg A. Distinct gene expression signatures in human embryonic stem cells differentiated towards definitive endoderm at single-cell level. Methods. 2013;59(1):59–70.

    Article  CAS  PubMed  Google Scholar 

  58. McGrath KE, Koniski AD, Maltby KM, McGann JK, Palis J. Embryonic expression and function of the chemokine SDF-1 and its receptor, CXCR4. Dev Biol. 1999;213(2):442–56.

    Article  CAS  PubMed  Google Scholar 

  59. Inui M, Fukui A, Ito Y, Asashima M. Xapelin and Xmsr are required for cardiovascular development in Xenopus laevis. Dev Biol. 2006;298(1):188–200.

    Article  CAS  PubMed  Google Scholar 

  60. Sağraç D, Şişli HB, Doğan A. Apelin receptor signaling during mesoderm development. Cell Biol Transl Med. 2020;10:1–15.

    Google Scholar 

  61. Geary L, LaBonne C. FGF mediated MAPK and PI3K/Akt Signals make distinct contributions to pluripotency and the establishment of neural crest. Elife. 2018;7:e33845.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Sittewelle M, Monsoro-Burq AH. AKT signaling displays multifaceted functions in neural crest development. Dev Biol. 2018;444:S144–55.

    Article  CAS  PubMed  Google Scholar 

  63. Liu Y, Kaneda R, Leja TW, Subkhankulova T, Tolmachov O, Minchiotti G, Schwartz RJ, Barahona M, Schneider MD. Hhex and Cer1 mediate the Sox17 pathway for cardiac mesoderm formation in embryonic stem cells. Stem Cells. 2014;32(6):1515–26.

    Article  CAS  PubMed  Google Scholar 

  64. Howard L, Mackenzie RM, Pchelintsev NA, McBryan T, McClure JD, McBride MW, Kane NM, Adams PD, Milligan G, Baker AH. Profiling of transcriptional and epigenetic changes during directed endothelial differentiation of human embryonic stem cells identifies FOXA2 as a marker of early mesoderm commitment. Stem Cell Res Ther. 2013;4(2):1–7.

    Article  Google Scholar 

  65. Kaiser S, Hackanson B, Follo M, Mehlhorn A, Geiger K, Ihorst G, Kapp U. BM cells giving rise to MSC in culture have a heterogeneous CD34 and CD45 phenotype. Cytotherapy. 2007;9(5):439–50.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This study was funded by TÜBİTAK 2232 International Fellowship for Outstanding Researchers Program (Project No: 118C186). The study was also supported by Yeditepe University. Ayşegül Doğan was supported by Turkish Academy of Sciences Outstanding Young Scientists Award (TÜBA-GEBİP 2020). We would like to thank Derya Sağraç for her help during immunofluorescence analysis. We would also like to thank Dr. Murat Kasap from Kocaeli University for his help during LC/MS analysis. Authors declare no conflict of interest.

Funding

This study was funded by TÜBİTAK 2232 International Fellowship for Outstanding Researchers Program (Project No. 118C186). The study was also supported by Yeditepe University. Ayşegül Doğan was supported by Turkish Academy of Sciences Outstanding Young Scientists Award (TÜBA-GEBİP 2020).

Author information

Authors and Affiliations

Authors

Contributions

AD: designed the whole project and experimental plan. Material preparation, data collection, and analysis were performed by HBŞ, SŞ, TBH, EB, and AD. Data interpretation was conducted by HBŞ and checked by AD. The first draft of the manuscript was written by HBŞ and AD. All authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Ayşegül Doğan.

Ethics declarations

Conflict of interest

The authors have no relevant financial or non-financial interests to disclose.

Additional information

Publisher's Note

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

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 Supplementary Video 1. Migration of Wild Type cell (R1)-derived EBs during mesoderm differentiation. (AVI 14753 KB)

Supplementary file2 Supplementary Video 2. Migration of Wild Type cell-derived EBs in the presence of Apelin-13 during mesoderm differentiation. (AVI 21286 KB)

Supplementary file3 Supplementary Video 3. Migration of Apelin KO cell-derived EBs during mesoderm differentiation. (AVI 14886 KB)

Supplementary file4 Supplementary Video 4. Migration of Aplnr KO cell-derived EBs during mesoderm differentiation. (AVI 15084 KB)

Supplementary file5 Supplementary Video 5. Migration of Elabela KO cell-derived EBs during mesoderm differentiation. (AVI 16037 KB)

Supplementary file6 Supplementary Video 6. Migration of R1 cell-derived EBs after Control shRNA-mediated silencing during mesoderm differentiation. (AVI 21995 KB)

Supplementary file7 Supplementary Video 7. Migration of R1 cell-derived EBs after shRNA-mediated silencing of Apelin during mesoderm differentiation. (AVI 18157 KB)

Supplementary file8 Supplementary Video 8. Migration of R1 cell-derived EBs after shRNA-mediated silencing of Aplnr during mesoderm differentiation. (AVI 24308 KB)

Supplementary file9 Supplementary Video 9. Migration of R1 cell-derived EBs after Control siRNA-mediated silencing during mesoderm differentiation. (AVI 14500 KB)

Supplementary file10 Supplementary Video 10. Migration of R1 cell-derived EBs after siRNA-mediated silencing of Apelin during mesoderm differentiation. (AVI 16868 KB)

Supplementary file11 Supplementary Video 11. Migration of R1 cell-derived EBs after siRNA-mediated silencing of Aplnr during mesoderm differentiation. (AVI 14156 KB)

13577_2023_861_MOESM12_ESM.tif

Supplementary file12 Supplementary Figure 1. Effect of knocking out Apelin, Aplnr and Elabela on migration of EBs during mesoderm differentiation of mESCs. (A) CRISPR constructs, carrying the gRNA scaffolds of Aplnr signaling components, to generate Apelin KO, Aplnr KO and Elabela KO cell lines. (B) The protocol for mesoderm differentiation of mESC. EBs, formed from mESCs, were transferred to Matrigel–Gelatin coated surface at Day2 and migration of the EBs were observed until Day 6. Apelin-13 or ML233 was optionally added at Day 2 to activate Aplnr signaling. (C) Morphological pictures of the EBs were taken at Day2, Day3 and Day6 for wild type cells. (D) Total area, core area and migration area (total area-core area) as well as core intensity and migration intensity were measured at Day 2, Day 3 and Day 6 and visualized as a heat map for Control, Apelin-13 and ML233 treated wild type EBs and (E) scattered line graphs of migration area and migration intensity as well as total migration area and total migration intensity between D6 and D2 were drawn. (F) Morphological pictures of the EBs were taken at Day2, Day3 and Day6 for Apelin KO cells. (G) Total area, core area and migration area (total area-core area) as well as core intensity and migration intensity were measured at Day 2, Day 3 and Day 6 and visualized as a heat map for Control, Apelin-13 and ML233 treated Apelin KO EBs and (H) scattered line graphs of migration area and migration intensity as well as total migration area and total migration intensity between D6 and D2 were drawn. (I) Morphological pictures of the EBs were taken at Day2, Day3 and Day6 for Elabela KO cells. (J) Total area, core area and migration area (total area-core area) as well as core intensity and migration intensity were measured at Day 2, Day 3 and Day 6 and visualized as a heat map for Control, Apelin-13 and ML233 treated Elabela KO EBs and (K) scattered line graphs of migration area and migration intensity as well as total migration area and total migration intensity between D6 and D2 were drawn. (L) Morphological pictures of the EBs were taken at Day2, Day3 and Day6 for Aplnr KO cells. (M) Total area, core area and migration area (total area-core area) as well as core intensity and migration intensity were measured at Day 2, Day 3 and Day 6 and visualized as a heat map for Control, Apelin-13 and ML233 treated Aplnr KO EBs and (N) scattered line graphs of migration area and migration intensity as well as total migration area and total migration intensity between D6 and D2 were drawn. (O) Bar graphs of total migration area (Day 6 Area - Day 2 Area) and (P) total migration intensity (Day 6 Intensity - Day 2 Intensity) compared to the ones of R1 were drawn. Scale bar: 50µm, 100µm. *P<0.05 (TIF 2260 KB)

13577_2023_861_MOESM13_ESM.tif

Supplementary file13 Supplementary Figure 2. Effect of shRNA and siRNA mediated silencing of Aplnr signaling on migration of EBs during mesoderm differentiation of mESCs. (A) Protocol for mesoderm differentiation of mESCs after lentiviral transduction of shApelin, shAplnr and shControl lentiviruses. EBs, formed from mESCs, were transferred to Matrigel–Gelatin coated surface at Day2 and incubated until Day 6. (B) Bar graph representation of qPCR analysis indicates relative gene expression of Apelin and Aplnr in the cells administered with the shRNA viruses. (C) Morphological pictures of the EBs were taken at Day2, Day3 and Day6. (D) Total area, core area and migration area (total area-core area) as well as core intensity and migration intensity were measured at Day 2, Day 3 and Day 6 and visualized as a heat map. (E) Scattered line graphs of migration area and migration intensity at Day 2, Day3 and Day6 as well as (F) bar graphs of total migration area and total migration intensity between D6 and D2 were drawn. (G) Protocol for mesoderm differentiation of mESCs after siRNA treatment. (H) Bar graph representation of qPCR analysis indicates relative gene expression of Apelin and Aplnr in the cells treated with Apelin siRNA, Aplnr siRNA and Control siRNA. (I) Morphological pictures of the EBs were taken at Day2, Day3 and Day6. (J) Total area, core area and migration area (total area-core area) as well as core intensity and migration intensity were measured at Day 2, Day 3 and Day 6 and visualized as a heat map. (K) Scattered line graphs of migration area and migration intensity at Day 2, Day3 and Day6 as well as (L) bar graphs of total migration area and total migration intensity between D6 and D2 were drawn for the siRNA treated EBs. Scale bar: 50µm, 100µm. *P<0.05 (TIF 1823 KB)

13577_2023_861_MOESM14_ESM.tif

Supplementary file14 Supplementary Figure 3. Effect of medium ingredients on Aplnr expression and migration of mESCs-derived EBs during mesoderm differentiation. (A) Medium supplements for each media options and timeline for mesoderm differentiation of R1 mESCs were shown. mESCs were used at Day0 to form EBs. The EBs were transferred to Matrigel–Gelatin coated surface at Day2 and their migration were observed until Day 6. (B) Morphological pictures of the EBs were taken at Day2, Day3 and Day6. (C) Total area, core area and migration area (total area-core area) as well as core intensity and migration intensity were measured at Day 2, Day 3 and Day 6 and visualized as a heat map. (D) Scattered line graphs of migration area and migration intensity at Day 2, Day3 and Day6 as well as bar graphs of total migration area and total migration intensity between D6 and D2 were drawn. (E) Heatmap of relative gene expression for neural rosette markers. ND: not detected. (F) Detection of Aplnr expression by flow cytometry at Day 2, Day 3 and Day 6 EBs. (G) Percentage of Aplnr positive cells in the populations were shown in the graph. Scale bar: 50µm, 100µm. *P<0.05 (TIF 2020 KB)

13577_2023_861_MOESM15_ESM.tif

Supplementary file15 Supplementary Figure 4. Effect of CRISPR-Cas9 mediated KO of Aplnr signaling components on mesoderm commitment of mESC-derived EBs during mesoderm differentiation. (A) The EBs were fixed using 4% paraformaldehyde at the end of Day3 and they were analyzed for protein expression of the Apelin, Aplnr, ectoderm marker protein, Sox2, the related pathway proteins, Akt and mTOR, MAPK pathway proteins involved in mesoderm commitment, Mek and Erk1/2, Pdgfrα, which guides migration of mesoderm cells, and mesoderm markers, Flk1 and MyoD. DAPI was used to stain nuclei of the cells. (B) IF analysis and the intensity of the expression as a heat map were shown. (C) Heatmap of relative gene expressions. Scale bar: 50µm. (TIF 5192 KB)

13577_2023_861_MOESM16_ESM.tif

Supplementary file16 Supplementary Figure 5. Effect of silencing Aplnr signaling components, via shRNA delivery on mesoderm commitment of mESC-derived EBs during mesoderm differentiation. (A) The EBs were fixed using 4% paraformaldehyde at the end of Day3 and they were analyzed for protein expression of the Apelin, Aplnr, ectoderm marker protein, Sox2, the related pathway proteins, Akt and mTOR, MAPK pathway proteins involved in mesoderm commitment, Mek and Erk1/2, Pdgfrα, which guides migration of mesoderm cells, and mesoderm markers, Flk1 and MyoD. DAPI was used to stain nuclei of the cells. (B) IF analysis and the intensity of the expression as a heat map were shown. (C) Heatmap of relative gene expressions. ND: not detected. Scale bar: 50µm. (TIF 3342 KB)

13577_2023_861_MOESM17_ESM.tif

Supplementary file17 Supplementary Figure 6. Effect of silencing Aplnr signaling components, via siRNA delivery on mesoderm commitment of mESC-derived EBs during mesoderm differentiation. (A) The EBs were fixed using 4% paraformaldehyde at the end of Day3 and they were analyzed for protein expression of the Apelin, Aplnr, ectoderm marker protein, Sox2, the related pathway proteins, Akt and mTOR, MAPK pathway proteins involved in mesoderm commitment, Mek and Erk1/2, Pdgfrα, which guides migration of mesoderm cells, and mesoderm markers, Flk1 and MyoD. DAPI was used to stain nuclei of the cells. (B) IF analysis and the intensity of the expression as a heat map were shown. (C) Heatmap of relative gene expressions. Scale bar: 50µm. (TIF 2506 KB)

13577_2023_861_MOESM18_ESM.tif

Supplementary file18 Supplementary Figure 7. Antibody array profile of mESC derived EBs treated with Apelin-13. (A) Membranes of the antibody array. Mesoderm related proteins were selected with black boxes on the membranes. (B) Hierarchical cluster analysis and heat map representation of proteins from Control and Apelin-13-treated EBs at Day 6. (C) Correlation coefficient matrix of the proteins. (D) Principal component analysis (PCA) of the cells. (E) A scattered graph indicating relative dot intensity for Control and Apelin-13-treated groups. (TIF 1632 KB)

13577_2023_861_MOESM19_ESM.tif

Supplementary file19 Supplementary Figure 8. Antibody array profile of mESC derived EBs when Aplnr was silenced. (A) Membranes of the antibody array. Mesoderm related proteins were selected with black boxes on the membranes. (B) Hierarchical cluster analysis and heat map representation of proteins from shControl and shAplnr EBs at Day 6. (C) Correlation coefficient matrix of the proteins. (D) Principal component analysis (PCA) of the cells. (E) A scattered graph indicating relative dot intensity for shControl and shAplnr groups. (TIF 1395 KB)

13577_2023_861_MOESM20_ESM.tif

Supplementary file20 Supplementary Figure 9. Optimization of MSC differentiation protocol from mESCs. (A) Protocols and timeline for MSC differentiation from mESCs. Standard protocol, mESC protocol and FCS protocol were applied. (B) Morphological pictures of R1 EBs at Day 2, Day 3, Day 6, Day 8 and Day 10. (C) Heatmap of relative gene expression analysis. Scale Bar: 200µm. (TIF 1243 KB)

13577_2023_861_MOESM21_ESM.tif

Supplementary file21 Supplementary Figure 10. Optimization of Apelin-13 addition to the MSC differentiation media. (A) Protocols and timelines for Apelin-13 addition to MSC differentiation protocol. (B) Morphological pictures of R1 EBs treated with the media options at Day 2, Day 3, Day 6, Day 8 and Day 10. (C) Flow cytometry analysis of R1 EBs at Day 10 for MSC markers and (D) its graphic representation indicating the positive cells for the markers. (E) A heat map representation of relative gene expressions for R1 EBs at Day 10. Scale Bar: 200µm. (TIF 2159 KB)

13577_2023_861_MOESM22_ESM.tif

Supplementary file22 Supplementary Figure 11. Effect of shRNA mediated silencing of Aplnr signaling components on MSC differentiation of R1 cells. (A) MSC differentiation protocol for Control, shApelin and shAplnr cells. (B) Morphological pictures of the cells taken at Day 2, Day 3, Day 6, Day 8 and Day 10 during MSC differentiation. (C) A heat map representation of relative gene expressions for the cells at Day 10. Scale Bar: 200µm. (TIF 1696 KB)

13577_2023_861_MOESM23_ESM.tif

Supplementary file23 Supplementary Figure 12. Effect of CRISPR-Cas9 mediated KO of Aplnr signaling components on MSC differentiation of R1 cells. (A) MSC differentiation protocol for wild type, Apelin KO, Aplnr KO and Elabela KO cells. (B) Morphological pictures of the cells taken at Day 2, Day 3, Day 6, Day 8 and Day 10 during MSC differentiation. (C) A heat map representation of relative gene expressions for the cells at Day 10. Scale Bar: 200µm. (TIF 2439 KB)

13577_2023_861_MOESM24_ESM.tif

Supplementary file24 Supplementary Figure 13. Effect of siRNA mediated silencing of Aplnr signaling components on MSC differentiation of R1 cells. (A) MSC differentiation protocol for Control, siApelin and siAplnr cells. (B) Morphological pictures of the cells taken at Day 2, Day 3, Day 6, Day 8 and Day 10 during MSC differentiation. (C) A heat map representation of relative gene expressions for the cells at Day 10. Scale Bar: 200µm. (TIF 1703 KB)

Supplementary file25 (DOCX 55 KB)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Şişli, H.B., Şenkal, S., Hayal, T.B. et al. Regulatory role of apelin receptor signaling in migration and differentiation of mouse embryonic stem cell-derived mesoderm cells and mesenchymal stem/stromal cells. Human Cell 36, 612–630 (2023). https://doi.org/10.1007/s13577-023-00861-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13577-023-00861-2

Keywords

Navigation