Browse > Article
http://dx.doi.org/10.1038/s12276-018-0185-z

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))
Publication Information
Experimental and Molecular Medicine / v.50, no.12, 2018 , pp. 5.1-5.15 More about this Journal
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
Citations & Related Records
연도 인용수 순위
  • Reference
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).   DOI
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).   DOI
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).   DOI
5 Ali, N. et al. Regulatory T cells in skin facilitate epithelial stem cell differentiation. Cell 169, 1119-1129 (2017).   DOI
6 Hoeck, J. D. et al. Stem cell plasticity enables hair regeneration following Lgr5 +cell loss. Nat. Cell Biol. 19, 666-676 (2017).   DOI
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).   DOI
8 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).   DOI
9 Chase, H. B. Growth of the hair. Physiol. Rev. 34, 113-126 (1954).   DOI
10 Dong, L. et al. Treatment of MSCs with Wnt1a-conditioned medium activates DP cells and promotes hair follicle regrowth. Sci. Rep. 4, 5432 (2014).
11 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).   DOI
12 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).   DOI
13 Ito, K. & Suda, T. Metabolic requirements for the maintenance of self-renewing stem cells. Nat. Rev. Mol. Cell Biol. 15, 243-256 (2014).   DOI
14 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).
15 Pryor, R. & Cabreiro, F. Repurposingmetformin: an old drug with new tricks in its binding pockets. Biochem. J. 471, 307-322 (2015).   DOI
16 Andrzejewski, S., Gravel, S. P., Pollak, M. & St-Pierre, J. Metformin directly acts on mitochondria to alter cellular bioenergetics. Cancer Metab. 2, 12 (2014).   DOI
17 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).   DOI
18 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).   DOI
19 Shyh-Chang, N. et al. Lin28 enhances tissue repair by reprogramming cellular metabolism. Cell 155, 778-792 (2013).   DOI
20 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).   DOI
21 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).   DOI
22 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).   DOI
23 Hamanaka, R. B. et al. Mitochondrial reactive oxygen species promote epidermal differentiation and hair follicle development. Sci. Signal. 6, ra8 (2013).   DOI
24 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).   DOI
25 Lane, S. W., Williams, D. A. & Watt, F. M. Modulating the stem cell niche for tissue regeneration. Nat. Biotechnol. 32, 795-803 (2014).   DOI
26 Shaw, R. J. AMPK keeps tumor cells from starving to death. Cell. Stem. Cell. 17, 503-504 (2015).   DOI
27 Rodriguez-Colman, M. J. et al. Interplay between metabolic identities in the intestinal crypt supports stem cell function. Nature 543, 424-427 (2017).   DOI
28 Shyh-Chang, N., Daley, G. Q. & Cantley, L. C. Stem cell metabolism in tissue development and aging. Development 140, 2535-2547 (2013).   DOI
29 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).   DOI
30 Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663-676 (2006).   DOI
31 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).   DOI
32 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).   DOI
33 Folmes, C. D. et al. Somatic oxidative bioenergetics transitions into pluripotency-dependent glycolysis to facilitate nuclear reprogramming. Cell. Metab. 14, 264-271 (2011).   DOI
34 Son, M. J. et al. Mitofusins deficiency elicits mitochondrial metabolic reprogramming to pluripotency. Cell Death Differ. 22, 1957-1969 (2015).   DOI
35 Ocampo, A. et al. In vivo amelioration of age-associated hallmarks by partial reprogramming. Cell 167, 1719-1733 e12 (2016).   DOI
36 Yu, J. et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 1917-1920 (2007).   DOI
37 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).   DOI
38 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).   DOI
39 Martin-Montalvo, A. et al. Metformin improves healthspan and lifespan in mice. Nat. Commun. 4, 2192 (2013).   DOI
40 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).   DOI
41 Theunissen, T. W. & Jaenisch, R. Molecular control of induced pluripotency. Cell. Stem. Cell. 14, 720-734 (2014).   DOI
42 Son, M. J. et al. Nicotinamide overcomes pluripotency deficits and reprogramming barriers. Stem Cells 31, 1121-1135 (2013).   DOI