DOI QR코드

DOI QR Code

A novel and safe small molecule enhances hair follicle regeneration by facilitating metabolic reprogramming

  • Son, Myung Jin (Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB)) ;
  • Jeong, Jae Kap (HanAll Biopharma) ;
  • Kwon, Youjeong (Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB)) ;
  • Ryu, Jae-Sung (Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB)) ;
  • Mun, Seon Ju (Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB)) ;
  • Kim, Hye Jin (Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB)) ;
  • Kim, Sung-wuk (HanAll Biopharma) ;
  • Yoo, Sanghee (ImmunoMet Therapeutics Inc., JLABS at Texas Medical Center) ;
  • Kook, Jiae (ImmunoMet Therapeutics Inc., JLABS at Texas Medical Center) ;
  • Lee, Hongbum (ImmunoMet Therapeutics Inc., JLABS at Texas Medical Center) ;
  • Kim, Janghwan (Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB)) ;
  • Chung, Kyung-Sook (Department of Functional Genomics, Korea University of Science & Technology (UST))
  • Received : 2018.04.10
  • Accepted : 2018.09.11
  • Published : 2018.12.30

Abstract

Targeting hair follicle regeneration has been investigated for the treatment of hair loss, and fundamental studies investigating stem cells and their niche have been described. However, knowledge of stem cell metabolism and the specific regulation of bioenergetics during the hair regeneration process is currently insufficient. Here, we report the hair regrowth-promoting effect of a newly synthesized novel small molecule, IM176OUT05 (IM), which activates stem cell metabolism. IM facilitated stemness induction and maintenance during an induced pluripotent stem cell generation process. IM treatment mildly inhibited mitochondrial oxidative phosphorylation and concurrently increased glycolysis, which accelerated stemness induction during the early phase of reprogramming. More importantly, the topical application of IM accelerated hair follicle regeneration by stimulating the progression of the hair follicle cycle to the anagen phase and increased the hair follicle number in mice. Furthermore, the stem cell population with a glycolytic metabotype appeared slightly earlier in the IM-treated mice. Stem cell and niche signaling involved in the hair regeneration process was also activated by the IM treatment during the early phase of hair follicle regeneration. Overall, these results show that the novel small molecule IM promotes tissue regeneration, specifically in hair regrowth, by restructuring the metabolic configuration of stem cells.

Keywords

Acknowledgement

Supported by : KHIDI, National Research Foundation of Korea

References

  1. Muller-Rover, S. et al. A comprehensive guide for the accurate classification of murine hair follicles in distinct hair cycle stages. J. Invest. Dermatol. 117, 3-15 (2001). https://doi.org/10.1046/j.0022-202x.2001.01377.x
  2. Genander, M. et al. BMP signaling and its pSMAD1/5 target genes differentially regulate hair follicle stem cell lineages. Cell. Stem. Cell. 15, 619-633 (2014). https://doi.org/10.1016/j.stem.2014.09.009
  3. Fuchs, E. Epithelial skin biology: three decades of developmental biology, a hundred questions answered and a thousand new ones to address. Curr. Top. Dev. Biol. 116, 357-374 (2016).
  4. Jaks, V. et al. Lgr5 marks cycling, yet long-lived, hair follicle stem cells. Nat. Genet. 40, 1291-1299 (2008). https://doi.org/10.1038/ng.239
  5. Ali, N. et al. Regulatory T cells in skin facilitate epithelial stem cell differentiation. Cell 169, 1119-1129 (2017). https://doi.org/10.1016/j.cell.2017.05.002
  6. Hoeck, J. D. et al. Stem cell plasticity enables hair regeneration following Lgr5 +cell loss. Nat. Cell Biol. 19, 666-676 (2017). https://doi.org/10.1038/ncb3535
  7. Plikus, M. V. et al. Local circadian clock gates cell cycle progression of transient amplifying cells during regenerative hair cycling. Proc. Natl. Acad. Sci. USA 110, E2106-E2115 (2013). https://doi.org/10.1073/pnas.1215935110
  8. Shyh-Chang, N. et al. Lin28 enhances tissue repair by reprogramming cellular metabolism. Cell 155, 778-792 (2013). https://doi.org/10.1016/j.cell.2013.09.059
  9. Kloepper, J. E. et al. Mitochondrial function in murine skin epithelium is crucial for hair follicle morphogenesis and epithelial-mesenchymal interactions. J. Invest. Dermatol. 135, 679-689 (2015). https://doi.org/10.1038/jid.2014.475
  10. Williams, R., Philpott, M. P. & Kealey, T. Metabolism of freshly isolated human hair follicles capable of hair elongation: a glutaminolytic, aerobic glycolytic tissue. J. Invest. Dermatol. 100, 834-840 (1993). https://doi.org/10.1111/1523-1747.ep12476744
  11. Kealey, T., Williams, R. & Philpott, M. P. The human hair follicle engages in glutaminolysis and aerobic glycolysis: implications for skin, splanchnic and neoplastic metabolism. Skin. Pharmacol. 7, 41-46 (1994). https://doi.org/10.1159/000211272
  12. Tang, Y. et al. Mitochondrial aerobic respiration is activated during hair follicle stem cell differentiation, and its dysfunction retards hair regeneration. PeerJ 4, e1821 (2016). https://doi.org/10.7717/peerj.1821
  13. Hamanaka, R. B. et al. Mitochondrial reactive oxygen species promote epidermal differentiation and hair follicle development. Sci. Signal. 6, ra8 (2013). https://doi.org/10.1126/scisignal.6305er8
  14. Shyh-Chang, N., Daley, G. Q. & Cantley, L. C. Stem cell metabolism in tissue development and aging. Development 140, 2535-2547 (2013). https://doi.org/10.1242/dev.091777
  15. Vidali, S. et al. Hypothalamic-pituitary-thyroid axis hormones stimulate mitochondrial function and biogenesis in human hair follicles. J. Invest. Dermatol. 134, 33-42 (2014). https://doi.org/10.1038/jid.2013.286
  16. Lemasters, J. J. et al. Compartmentation of mitochondrial and oxidative metabolism in growing hair follicles: a ring of fire. J. Invest. Dermatol. 137, 1434-1444 (2017). https://doi.org/10.1016/j.jid.2017.02.983
  17. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663-676 (2006). https://doi.org/10.1016/j.cell.2006.07.024
  18. Theunissen, T. W. & Jaenisch, R. Molecular control of induced pluripotency. Cell. Stem. Cell. 14, 720-734 (2014). https://doi.org/10.1016/j.stem.2014.05.002
  19. Folmes, C. D. et al. Somatic oxidative bioenergetics transitions into pluripotency-dependent glycolysis to facilitate nuclear reprogramming. Cell. Metab. 14, 264-271 (2011). https://doi.org/10.1016/j.cmet.2011.06.011
  20. Son, M. J. et al. Mitofusins deficiency elicits mitochondrial metabolic reprogramming to pluripotency. Cell Death Differ. 22, 1957-1969 (2015). https://doi.org/10.1038/cdd.2015.43
  21. Ocampo, A. et al. In vivo amelioration of age-associated hallmarks by partial reprogramming. Cell 167, 1719-1733 e12 (2016). https://doi.org/10.1016/j.cell.2016.11.052
  22. Yu, J. et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 1917-1920 (2007). https://doi.org/10.1126/science.1151526
  23. Son, M. J., Jeong, B. R., Kwon, Y. & Cho, Y. S. Interference with the mitochondrial bioenergetics fuels reprogramming to pluripotency via facilitation of the glycolytic transition. Int. J. Biochem. Cell. Biol. 45, 2512-2518 (2013). https://doi.org/10.1016/j.biocel.2013.07.023
  24. Dadwal, P. et al. Activating endogenous neural precursor cells using metformin leads to neural repair and functional recovery in a model of childhood brain injury. Stem Cell Rep. 5, 166-173 (2015). https://doi.org/10.1016/j.stemcr.2015.06.011
  25. Wang, J. et al. Metformin activates an atypical PKC-CBP pathway to promote neurogenesis and enhance spatial memory formation. Cell. Stem. Cell. 11, 23-35 (2012). https://doi.org/10.1016/j.stem.2012.03.016
  26. Martin-Montalvo, A. et al. Metformin improves healthspan and lifespan in mice. Nat. Commun. 4, 2192 (2013). https://doi.org/10.1038/ncomms3192
  27. El-Mir, M. Y. et al. Dimethylbiguanide inhibits cell respiration via an indirect effect targeted on the respiratory chain complex I. J. Biol. Chem. 275, 223-228 (2000). https://doi.org/10.1074/jbc.275.1.223
  28. Son, M. J. et al. Nicotinamide overcomes pluripotency deficits and reprogramming barriers. Stem Cells 31, 1121-1135 (2013). https://doi.org/10.1002/stem.1368
  29. Son, M. J. et al. Upregulation of mitochondrial NAD+levels impairs the clonogenicity of SSEA1+glioblastoma tumor-initiating cells. Exp. Mol. Med. 49, e344 (2017). https://doi.org/10.1038/emm.2017.74
  30. Chase, H. B. Growth of the hair. Physiol. Rev. 34, 113-126 (1954). https://doi.org/10.1152/physrev.1954.34.1.113
  31. Dong, L. et al. Treatment of MSCs with Wnt1a-conditioned medium activates DP cells and promotes hair follicle regrowth. Sci. Rep. 4, 5432 (2014).
  32. Enshell-Seijffers, D., Lindon, C., Kashiwagi, M. & Morgan, B. A. beta-catenin activity in the dermal papilla regulates morphogenesis and regeneration of hair. Dev. Cell. 18, 633-642 (2010). https://doi.org/10.1016/j.devcel.2010.01.016
  33. Andl, T., Reddy, S. T., Gaddapara, T. & Millar, S. E. WNT signals are required for the initiation of hair follicle development. Dev. Cell. 2, 643-653 (2002). https://doi.org/10.1016/S1534-5807(02)00167-3
  34. Ito, K. & Suda, T. Metabolic requirements for the maintenance of self-renewing stem cells. Nat. Rev. Mol. Cell Biol. 15, 243-256 (2014). https://doi.org/10.1038/nrm3772
  35. Becker, M. L. et al. Genetic variation in the organic cation transporter 1 is associated with metformin response in patients with diabetes mellitus. Pharm. J. 9, 242-247 (2009).
  36. Pryor, R. & Cabreiro, F. Repurposingmetformin: an old drug with new tricks in its binding pockets. Biochem. J. 471, 307-322 (2015). https://doi.org/10.1042/BJ20150497
  37. Andrzejewski, S., Gravel, S. P., Pollak, M. & St-Pierre, J. Metformin directly acts on mitochondria to alter cellular bioenergetics. Cancer Metab. 2, 12 (2014). https://doi.org/10.1186/2049-3002-2-12
  38. Fu, X., Zhu, M. J., Dodson, M. V. & Du, M. AMP-activated protein kinase stimulates Warburg-like glycolysis and activation of satellite cells during muscle regeneration. J. Biol. Chem. 290, 26445-26456 (2015). https://doi.org/10.1074/jbc.M115.665232
  39. Chandel, N. S., Jasper, H., Ho, T. T. & Passegue, E. Metabolic regulation of stem cell function in tissue homeostasis and organismal ageing. Nat. Cell Biol. 18, 823-832 (2016). https://doi.org/10.1038/ncb3385
  40. Lane, S. W., Williams, D. A. & Watt, F. M. Modulating the stem cell niche for tissue regeneration. Nat. Biotechnol. 32, 795-803 (2014). https://doi.org/10.1038/nbt.2978
  41. Shaw, R. J. AMPK keeps tumor cells from starving to death. Cell. Stem. Cell. 17, 503-504 (2015). https://doi.org/10.1016/j.stem.2015.10.007
  42. Rodriguez-Colman, M. J. et al. Interplay between metabolic identities in the intestinal crypt supports stem cell function. Nature 543, 424-427 (2017). https://doi.org/10.1038/nature21673

Cited by

  1. Bioactive Molecules for Skin Repair and Regeneration: Progress and Perspectives vol.2019, pp.None, 2018, https://doi.org/10.1155/2019/6789823
  2. Androgenetic alopecia: combing the hair follicle signaling pathways for new therapeutic targets and more effective treatment options vol.23, pp.9, 2019, https://doi.org/10.1080/14728222.2019.1659779
  3. Restoring aged stem cell functionality: Current progress and future directions vol.38, pp.9, 2020, https://doi.org/10.1002/stem.3234
  4. Why All the Fuss about Oxidative Phosphorylation (OXPHOS)? vol.63, pp.23, 2018, https://doi.org/10.1021/acs.jmedchem.0c01013
  5. Small Molecule Treatments Improve Differentiation Potential of Human Amniotic Fluid Stem Cells vol.9, pp.None, 2018, https://doi.org/10.3389/fbioe.2021.623886
  6. Transdermal Drug Delivery for Hair Regrowth vol.18, pp.2, 2018, https://doi.org/10.1021/acs.molpharmaceut.0c00041
  7. Inhibition of pyruvate oxidation as a versatile stimulator of the hair cycle in models of alopecia vol.30, pp.4, 2021, https://doi.org/10.1111/exd.14307
  8. Long-Chain Free Fatty Acids Influence Lipid Accumulation, Lysosome Activation and Glycolytic Shift in Various Cells In Vitro vol.55, pp.4, 2021, https://doi.org/10.1134/s0026893321030146