DOI QR코드

DOI QR Code

Low-dose metronomic doxorubicin inhibits mobilization and differentiation of endothelial progenitor cells through REDD1-mediated VEGFR-2 downregulation

  • Park, Minsik (Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine) ;
  • Kim, Ji Yoon (Department of Anaesthesiology and Pain Medicine, Hanyang University Hospital) ;
  • Kim, Joohwan (Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine) ;
  • Lee, Jeong-Hyung (Department of Biochemistry, Kangwon National University) ;
  • Kwon, Young-Guen (Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University) ;
  • Kim, Young-Myeong (Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine)
  • Received : 2021.07.19
  • Accepted : 2021.08.10
  • Published : 2021.09.30

Abstract

Low-dose metronomic chemotherapy has been introduced as a less toxic and effective strategy to inhibit tumor angiogenesis, but its anti-angiogenic mechanism on endothelial progenitor cells (EPCs) has not been fully elucidated. Here, we investigated the functional role of regulated in development and DNA damage response 1 (REDD1), an endogenous inhibitor of mTORC1, in low-dose doxorubicin (DOX)-mediated dysregulation of EPC functions. DOX treatment induced REDD1 expression in bone marrow mononuclear cells (BMMNCs) and subsequently reduced mTORC1-dependent translation of endothelial growth factor (VEGF) receptor (Vegfr)-2 mRNA, but not that of the mRNA transcripts for Vegfr-1, epidermal growth factor receptor, and insulin-like growth factor-1 receptor. This selective event was a risk factor for the inhibition of BMMNC differentiation into EPCs and their angiogenic responses to VEGF-A, but was not observed in Redd1-deficient BMMNCs. Low-dose metronomic DOX treatment reduced the mobilization of circulating EPCs in B16 melanoma-bearing wild-type but not Redd1-deficient mice. However, REDD1 overexpression inhibited the differentiation and mobilization of EPCs in both wild-type and Redd1-deficient mice. These data suggest that REDD1 is crucial for metronomic DOX-mediated EPC dysfunction through the translational repression of Vegfr-2 transcript, providing REDD1 as a potential therapeutic target for the inhibition of tumor angiogenesis and tumor progression.

Keywords

Acknowledgement

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIP) (No. 2017R1A2B3004565).

References

  1. Shibuya M (2011) Vascular endothelial growth factor (VEGF) and its receptor (VEGFR) signaling in angiogenesis: a crucial target for anti- and pro-angiogenic therapies. Genes Cancer 2, 1097-1105 https://doi.org/10.1177/1947601911423031
  2. Beck B, Driessens G, Goossens S et al (2011) A vascular niche and a VEGF-Nrp1 loop regulate the initiation and stemness of skin tumours. Nature 478, 399-403 https://doi.org/10.1038/nature10525
  3. Kim JY and Kim YM (2019) Tumor endothelial cells as a potential target of metronomic chemotherapy. Arch Pharm Res 42, 1-13 https://doi.org/10.1007/s12272-018-01102-z
  4. Kerbel RS (2008) Tumor angiogenesis. N Engl J Med 358, 2039-2049 https://doi.org/10.1056/NEJMra0706596
  5. Stoelting S, Trefzer T, Kisro J, Steinke A, Wagner T and Peters SO (2008) Low-dose oral metronomic chemotherapy prevents mobilization of endothelial progenitor cells into the blood of cancer patients. In Vivo 22, 831-836
  6. Kerbel RS and Kamen BA (2004) The anti-angiogenic basis of metronomic chemotherapy. Nat Rev Cancer 4, 423-436 https://doi.org/10.1038/nrc1369
  7. Rasanen M, Degerman J, Nissinen TA et al (2016) VEGF-B gene therapy inhibits doxorubicin-induced cardiotoxicity by endothelial protection. Proc Natl Acad Sci U S A 113, 13144-13149 https://doi.org/10.1073/pnas.1616168113
  8. Park M, Kim J, Kim T et al (2021) REDD1 is a determinant of low-dose metronomic doxorubicin-elicited endothelial cell dysfunction through downregulation of VEGFR-2/3. Exp Mol Med (In press)
  9. DeYoung MP, Horak P, Sofer A, Sgroi D and Ellisen LW (2008) Hypoxia regulates TSC1/2-mTOR signaling and tumor suppression through REDD1-mediated 14-3-3 shuttling. Genes Dev 22, 239-251 https://doi.org/10.1101/gad.1617608
  10. Lee DK, Kim JH, Kim J et al (2018) REDD-1 aggravates endotoxin-induced inflammation via atypical NF-κB activation. FASEB J 32, 4585-4599 https://doi.org/10.1096/fj.201701436R
  11. Karar J and Maity A (2011) PI3K/AKT/mTOR Pathway in Angiogenesis. Front Mol Neurosci 4, 51 https://doi.org/10.3389/fnmol.2011.00051
  12. Guba M, von Breitenbuch P, Steinbauer M et al (2002) Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor. Nat Med 8, 128-135 https://doi.org/10.1038/nm0202-128
  13. Wang M, Xu Y, Wen GZ, Wang Q and Yuan SM (2019) Rapamycin suppresses angiogenesis and lymphangiogenesis in melanoma by downregulating VEGF-A/VEGFR-2 and VEGF-C/VEGFR-3 expression. Onco Targets Ther 12, 4643-4654 https://doi.org/10.2147/OTT.S205160
  14. Marumo T, Uchimura H, Hayashi M, Hishikawa K and Fujita T (2006) Aldosterone impairs bone marrow-derived progenitor cell formation. Hypertension 48, 490-496 https://doi.org/10.1161/01.HYP.0000235681.25685.cf
  15. Pitchford SC, Furze RC, Jones CP, Wengner AM and Rankin SM (2009) Differential mobilization of subsets of progenitor cells from the bone marrow. Cell Stem Cell 4, 62-72 https://doi.org/10.1016/j.stem.2008.10.017
  16. Aicher A, Heeschen C, Mildner-Rihm C et al (2003) Essential role of endothelial nitric oxide synthase for mobilization of stem and progenitor cells. Nat Med 9, 1370-1376 https://doi.org/10.1038/nm948
  17. Bertolini F, Paul S, Mancuso P, Monestiroli S, Gobbi A, Shaked Y and Kerbel RS (2003) Maximum tolerable dose and low-dose metronomic chemotherapy have opposite effects on the mobilization and viability of circulating endothelial progenitor cells. Cancer Res 63, 4342-4346
  18. Bocci G, Francia G, Man S, Lawler J and Kerbel RS (2003) Thrombospondin 1, a mediator of the antiangiogenic effects of low-dose metronomic chemotherapy. Proc Natl Acad Sci U S A 100, 12917-12922 https://doi.org/10.1073/pnas.2135406100
  19. Saif MW (2013) Anti-VEGF agents in metastatic colorectal cancer (mCRC): are they all alike? Cancer Manag Res 5, 103-115 https://doi.org/10.2147/CMAR.S45193
  20. Asahara T, Murohara T, Sullivan A et al (1997) Isolation of putative progenitor endothelial cells for angiogenesis. Science 275, 964-967 https://doi.org/10.1126/science.275.5302.964
  21. Asahara T, Masuda H, Takahashi T et al (1999) Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ Res 85, 221-228 https://doi.org/10.1161/01.RES.85.3.221
  22. Willett CG, Boucher Y, di Tomaso E et al (2004) Direct evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer. Nat Med 10, 145-147 https://doi.org/10.1038/nm988
  23. Murakami J, Li TS, Ueda K, Tanaka T and Hamano K (2009) Inhibition of accelerated tumor growth by blocking the recruitment of mobilized endothelial progenitor cells after chemotherapy. Int J Cancer 12, 1685-1692
  24. Sxton RA and Sabatini DM (2017) mTOR Signaling in growth, metabolism, and disease. Cell 169, 361-371
  25. Giraud S, Greco A, Brink M, Diaz JJ and Delafontaine P (2001) Translation initiation of the insulin-like growth factor I receptor mRNA is mediated by an internal ribosome entry site. J Biol Chem 276, 5668-5675 https://doi.org/10.1074/jbc.M005928200
  26. Chen TG, Chen JZ and Wang XX (2006) Effects of rapamycin on number activity and eNOS of endothelial progenitor cells from peripheral blood. Cell Prolif 39, 117-125 https://doi.org/10.1111/j.1365-2184.2006.00375.x