Thyroid hormone signaling controls hair follicle stem cell function

Constanza Contreras-Jurado, Corina Lorz, Laura García-Serrano, Jesus M Paramio, Ana Aranda, Constanza Contreras-Jurado, Corina Lorz, Laura García-Serrano, Jesus M Paramio, Ana Aranda

Abstract

Observations in thyroid patients and experimental animals show that the skin is an important target for the thyroid hormones. We previously showed that deletion in mice of the thyroid hormone nuclear receptors TRα1 and TRβ (the main thyroid hormone-binding isoforms) results in impaired epidermal proliferation, hair growth, and wound healing. Stem cells located at the bulges of the hair follicles are responsible for hair cycling and contribute to the regeneration of the new epidermis after wounding. Therefore a reduction in the number or function of the bulge stem cells could be responsible for this phenotype. Bulge cells show increased levels of epigenetic repressive marks, can retain bromodeoxyuridine labeling for a long time, and have colony-forming efficiency (CFE) in vitro. Here we demonstrate that mice lacking TRs do not have a decrease of the bulge stem cell population. Instead, they show an increase of label-retaining cells (LRCs) in the bulges and enhanced CFE in vitro. Reduced activation of stem cells leading to their accumulation in the bulges is indicated by a strongly reduced response to mobilization by 12-O-tetradecanolyphorbol-13-acetate. Altered function of the bulge stem cells is associated with aberrant activation of Smad signaling, leading to reduced nuclear accumulation of β-catenin, which is crucial for stem cell proliferation and mobilization. LRCs of TR-deficient mice also show increased levels of epigenetic repressive marks. We conclude that thyroid hormone signaling is an important determinant of the mobilization of stem cells out of their niche in the hair bulge. These findings correlate with skin defects observed in mice and alterations found in human thyroid disorders.

© 2015 Contreras-Jurado et al. This article is distributed by The American Society for Cell Biology under license from the author(s). Two months after publication it is available to the public under an Attribution–Noncommercial–Share Alike 3.0 Unported Creative Commons License (http://creativecommons.org/licenses/by-nc-sa/3.0).

Figures

FIGURE 1:
FIGURE 1:
TR inactivation does not lead to SC depletion in hair bulges. (A) Double immunofluorescence of SC markers keratin 15 (K15), CD34, and Sox9 in the bulges of dorsal skin of WT and TR KO mice. Nuclei were counterstained with DAPI (blue), and merge images are shown. (B) Confocal images of α6 integrin, CD34, and K15 expression in whole mounts of tail skin in both groups. (C) Transcript levels of the indicated SC markers determined in telogen epidermis of WT and TR KO animals. Results are expressed relative to the values obtained in WT mice. (D) Flow cytometry analysis of surface levels of CD34 and α6 integrin in WT and KO mice. Subpopulations of CD34+ cells expressing high and low levels of α6 integrin were calculated. Data are means ± SD. *p < 0.05, **p < 0.01.
FIGURE 2:
FIGURE 2:
LRC quantification in whole mounts of tail epidermis. (A) WT and TR KO neonatal mice (six mice/group) were injected with BrdU, and LRCs were identified by immunofluorescence after 75 d. (B) BrdU labeling intensity in the bulges of untreated WT and TR-deficient mice was quantified, and the number of LRCs with high, medium, and low label intensity is shown. High intensity represents nuclei in which the labeled area is >55 μm2; medium intensity, >42 but <55 μm2; and low intensity, >28 but <42 μm2. **p < 0.01.
FIGURE 3:
FIGURE 3:
Enhanced colony formation efficiency in TR-deficient mice (A) Representative images of CFE of keratinocytes isolated from adult WT and TR KO mice plated at two different concentrations (5 × 104 and 25 × 104 cells/well). (B) Quantification of the size and number of colonies obtained in both genotypes. Data are means ± SD. **p < 0.01.
FIGURE 4:
FIGURE 4:
LRCs in the bulges of TR KO mice are enriched in trimethylated histone 3 at lysines 9 and 27. (A) Neonatal mice were injected with BrdU, and after the chase period, whole mounts of tail epidermis from WT and TR KO mice were used for double immunofluorescence with H3K9me3 and BrdU antibodies. (B) Quantification of the label intensity of H3K9me3 in the bulges, hair germs, and LRCs (BrdU-positive cells in the bulges). (C) Representative images obtained as in A with BrdU and H3K27me3 antibodies. (D) Quantification of H3K27me3 in the bulges, hair germs, and LRCs. (E) Double immunofluorescence images of BrdU and acetyl histone 4 (AcH4). (F) Quantification of AcH4 in the bulges, hair germs, and LRCs of WT and TR KO mice. Data are means ± SE. *p < 0.05, **p < 0.01, and ***p < 0.001.
FIGURE 5:
FIGURE 5:
TR deletion blocks LRC mobilization. (A) Neonatal mice were labeled with BrdU, and 68 d later, tails were topically treated with TPA as described in Materials and Methods. Representative LRC images of BrdU immunofluorescence in whole mounts of tail epidermis from vehicle-treated (control) and TPA-treated WT and TR KO mice. (B) Number of LRCs in which the label occupied an area >28 μm2 was scored in control and TPA-treated mice. ANOVA followed by Bonferroni test was used to analyze differences among groups. (C) Percentage of LRCs mobilized upon TPA treatment in both genotypes. Data are means ± SD. **p < 0.01 ***p < 0.001.
FIGURE 6:
FIGURE 6:
Smad signaling is aberrantly activated in the follicles of TR KO mice. (A) Double immunofluorescence images of K14 and pSmad1,5,8 in the skin of control and TPA-treated WT and TR KO mice, showing increased Smad phosphorylation in the hair follicles of TR-deficient mice. (B) Double immunofluorescence of nuclear β-catenin and K5 in the same groups. Hair follicles are marked by a white line, and brackets indicate the location of the bulges (Bu) and the sebaceous gland (Sg). Bars, 150 μm.
FIGURE 7:
FIGURE 7:
Reduced expression of β-catenin targets in TR-deficient mice. (A) Transcript levels of the β-catenin target genes Cyclin D1 and c-Myc determined in epidermis of control and TPA-treated WT and TR KO animals. Results are expressed relative to the Gus mRNA levels. Data are means ± SD. **p < 0.01, ***p < 0.001. (B) Double immunofluorescence of K5 and activated c-Myc in the skin of control and TPA-treated WT and TR KO mice.
FIGURE 8:
FIGURE 8:
Reduced Smad signaling in the hair follicles of hyperthyroid mice. (A) Representative double immunofluorescence images of K14 and pSmad1,5,8 in the follicles of control mice and mice treated for 30 d with thyroid hormones (hyperthyroid). Arrows indicate the presence of cells positive for the phosphorylated Smad. (B) K5 and nuclear β-catenin detected by double immunofluorescence in control and hyperthyroid hair follicles. β-Catenin–positive cells are shown with arrows. Bu, bulge; SG, sebaceous gland. Bars,150 μm.

References

    1. Ahsan MK, Urano Y, Kato S, Oura H, Arase S. Immunohistochemical localization of thyroid hormone nuclear receptors in human hair follicles and in vitro effect of L-triiodothyronine on cultured cells of hair follicles and skin. J Med Invest. 1998;44:179–184.
    1. Arnold I, Watt FM. c-Myc activation in transgenic mouse epidermis results in mobilization of stem cells and differentiation of their progeny. Curr Biol. 2001;11:558–568.
    1. Barrandon Y, Green H. Three clonal types of keratinocyte with different capacities for multiplication. Proc Natl Acad Sci USA. 1987;84:2302–2306.
    1. Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, Wei G, Chepelev I, Zhao K. High-resolution profiling of histone methylations in the human genome. Cell. 2007;129:823–837.
    1. Benitah SA. Defining an epidermal stem cell epigenetic network. Nat Cell Biol. 2012;14:652–653.
    1. Billoni N, Buan B, Gautier B, Gaillard O, Mahe YF, Bernard BA. Thyroid hormone receptor beta1 is expressed in the human hair follicle. Br J Dermatol. 2000;142:645–652.
    1. Blanpain C, Fuchs E. Epidermal homeostasis: a balancing act of stem cells in the skin. Nat Rev Mol Cell Biol. 2009;10:207–217.
    1. Blanpain C, Lowry WE, Geoghegan A, Polak L, Fuchs E. Self-renewal, multipotency, and the existence of two cell populations within an epithelial stem cell niche. Cell. 2004;118:635–648.
    1. Botchkarev VA, Botchkareva NV, Roth W, Nakamura M, Chen LH, Herzog W, Lindner G, McMahon JA, Peters C, Lauster R, et al. Noggin is a mesenchymally derived stimulator of hair-follicle induction. Nat Cell Biol. 1999;1:158–164.
    1. Braun KM, Niemann C, Jensen UB, Sundberg JP, Silva-Vargas V, Watt FM. Manipulation of stem cell proliferation and lineage commitment: visualisation of label-retaining cells in wholemounts of mouse epidermis. Development. 2003;130:5241–5255.
    1. Claudinot S, Nicolas M, Oshima H, Rochat A, Barrandon Y. Long-term renewal of hair follicles from clonogenic multipotent stem cells. Proc Natl Acad Sci USA. 2005;102:14677–14682.
    1. Clevers H. Wnt/beta-catenin signaling in development and disease. Cell. 2006;127:469–480.
    1. Contreras-Jurado C, Garcia-Serrano L, Gomez-Ferreria M, Costa C, Paramio JM, Aranda A. The thyroid hormone receptors as modulators of skin proliferation and inflammation. J Biol Chem. 2011;286:24079–24088.
    1. Contreras-Jurado C, García-Serrano L, Martínez-Fernández M, Ruiz-Llorente L, Paramio JM, Aranda A. Impaired hair growth and wound healing in mice lacking thyroid hormone receptors. PLoS One. 2014;9:e108137.
    1. Cotsarelis G, Sun TT, Lavker RM. Label-retaining cells reside in the bulge area of pilosebaceous unit: implications for follicular stem cells, hair cycle, and skin carcinogenesis. Cell. 1990;61:1329–1337.
    1. Dentice M, Marsili A, Ambrosio R, Guardiola O, Sibilio A, Paik JH, Minchiotti G, DePinho RA, Fenzi G, Larsen PR, Salvatore D. The FoxO3/type 2 deiodinase pathway is required for normal mouse myogenesis and muscle regeneration. J Clin Invest. 2010;120:4021–4030.
    1. Freinkel RK, Freinkel N. Hair growth and alopecia in hypothyroidism. Arch Dermatol. 1972;106:349–352.
    1. Frye M, Fisher AG, Watt FM. Epidermal stem cells are defined by global histone modifications that are altered by Myc-induced differentiation. PLoS One. 2007;2:e763.
    1. Fuchs E, Chen T. A matter of life and death: self-renewal in stem cells. EMBO Rep. 2013;14:39–48.
    1. Garcia-Serrano L, Gomez-Ferreria MA, Contreras-Jurado C, Segrelles C, Paramio JM, Aranda A. The thyroid hormone receptors modulate the skin response to retinoids. PLoS One. 2011;6:e23825.
    1. Gothe S, Wang Z, Ng L, Kindblom JM, Barros AC, Ohlsson C, Vennstrom B, Forrest D. Mice devoid of all known thyroid hormone receptors are viable but exhibit disorders of the pituitary-thyroid axis, growth, and bone maturation. Genes Dev. 1999;13:1329–1341.
    1. Greco V, Chen T, Rendl M, Schober M, Pasolli HA, Stokes N, Dela Cruz-Racelis J, Fuchs E. A two-step mechanism for stem cell activation during hair regeneration. Cell Stem Cell. 2009;4:155–169.
    1. Horsley V, Aliprantis AO, Polak L, Glimcher LH, Fuchs E. NFATc1 balances quiescence and proliferation of skin stem cells. Cell. 2008;132:299–310.
    1. Huelsken J, Vogel R, Erdmann B, Cotsarelis G, Birchmeier W. beta-Catenin controls hair follicle morphogenesis and stem cell differentiation in the skin. Cell. 2001;105:533–545.
    1. Ito M, Liu Y, Yang Z, Nguyen J, Liang F, Morris RJ, Cotsarelis G. Stem cells in the hair follicle bulge contribute to wound repair but not to homeostasis of the epidermis. Nat Med. 2005;11:1351–1354.
    1. Jaks V, Barker N, Kasper M, van Es JH, Snippert HJ, Clevers H, Toftgard R. Lgr5 marks cycling, yet long-lived, hair follicle stem cells. Nat Genet. 2008;40:1291–1299.
    1. Jamora C, DasGupta R, Kocieniewski P, Fuchs E. Links between signal transduction, transcription and adhesion in epithelial bud development. Nature. 2003;422:317–322.
    1. Jones PH, Watt FM. Separation of human epidermal stem cells from transit amplifying cells on the basis of differences in integrin function and expression. Cell. 1993;73:713–724.
    1. Kandyba E, Leung Y, Chen YB, Widelitz R, Chuong CM, Kobielak K. Competitive balance of intrabulge BMP/Wnt signaling reveals a robust gene network ruling stem cell homeostasis and cyclic activation. Proc Natl Acad Sci USA. 2013;110:1351–1356.
    1. Kobayashi K, Rochat A, Barrandon Y. Segregation of keratinocyte colony-forming cells in the bulge of the rat vibrissa. Proc Natl Acad Sci USA. 1993;90:7391–7395.
    1. Kobielak K, Stokes N, de la Cruz J, Polak L, Fuchs E. Loss of a quiescent niche but not follicle stem cells in the absence of bone morphogenetic protein signaling. Proc Natl Acad Sci USA. 2007;104:10063–10068.
    1. Lemkine GF, Raj A, Alfama G, Turque N, Hassani Z, Alegria-Prevot O, Samarut J, Levi G, Demeneix BA. Adult neural stem cell cycling in vivo requires thyroid hormone and its alpha receptor. FASEB J. 2005;19:863–865.
    1. Levy V, Lindon C, Zheng Y, Harfe BD, Morgan BA. Epidermal stem cells arise from the hair follicle after wounding. FASEB J. 2007;21:1358–1366.
    1. Lopez-Juarez A, Remaud S, Hassani Z, Jolivet P, Pierre Simons J, Sontag T, Yoshikawa K, Price J, Morvan-Dubois G, Demeneix BA. Thyroid hormone signaling acts as a neurogenic switch by repressing Sox2 in the adult neural stem cell niche. Cell Stem Cell. 2012;10:531–543.
    1. Lorz C, Garcia-Escudero R, Segrelles C, Garin MI, Ariza JM, Santos M, Ruiz S, Lara MF, Martinez-Cruz AB, Costa C, et al. A functional role of RB-dependent pathway in the control of quiescence in adult epidermal stem cells revealed by genomic profiling. Stem Cell Rev. 2010;6:162–177.
    1. Lowry WE, Blanpain C, Nowak JA, Guasch G, Lewis L, Fuchs E. Defining the impact of beta-catenin/Tcf transactivation on epithelial stem cells. Genes Dev. 2005;19:1596–1611.
    1. Martinez-Iglesias O, Garcia-Silva S, Tenbaum SP, Regadera J, Larcher F, Paramio JM, Vennstrom B, Aranda A. Thyroid hormone receptor beta1 acts as a potent suppressor of tumor invasiveness and metastasis. Cancer Res. 2009;69:501–509.
    1. Messenger AG. Thyroid hormone and hair growth. Br J Dermatol. 2000;142:633–634.
    1. Moore RJ, Owens DM, Stamp G, Arnott C, Burke F, East N, Holdsworth H, Turner L, Rollins B, Pasparakis M, et al. Mice deficient in tumor necrosis factor-alpha are resistant to skin carcinogenesis. Nat Med. 1999;5:828–831.
    1. Morris RJ, Liu Y, Marles L, Yang Z, Trempus C, Li S, Lin JS, Sawicki JA, Cotsarelis G. Capturing and profiling adult hair follicle stem cells. Nat Biotechnol. 2004;22:411–417.
    1. Morris RJ, Potten CS. Highly persistent label-retaining cells in the hair follicles of mice and their fate following induction of anagen. J Invest Dermatol. 1999;112:470–475.
    1. Nowak JA, Polak L, Pasolli HA, Fuchs E. Hair follicle stem cells are specified and function in early skin morphogenesis. Cell Stem Cell. 2008;3:33–43.
    1. Oshima H, Rochat A, Kedzia C, Kobayashi K, Barrandon Y. Morphogenesis and renewal of hair follicles from adult multipotent stem cells. Cell. 2001;104:233–245.
    1. Pascual A, Aranda A. Thyroid hormone receptors, cell growth and differentiation. Biochim Biophys Acta. 2013;1830:3908–3916.
    1. Paus R. Exploring the “thyroid-skin connection”: concepts, questions, and clinical relevance. J Invest Dermatol. 2010;130:7–10.
    1. Plateroti M, Kress E, Mori JI, Samarut J. Thyroid hormone receptor alpha1 directly controls transcription of the beta-catenin gene in intestinal epithelial cells. Mol Cell Biol. 2006;26:3204–3214.
    1. Plikus MV, Mayer JA, de la Cruz D, Baker RE, Maini PK, Maxson R, Chuong CM. Cyclic dermal BMP signalling regulates stem cell activation during hair regeneration. Nature. 2008;451:340–344.
    1. Rhee H, Polak L, Fuchs E. Lhx2 maintains stem cell character in hair follicles. Science. 2006;312:1946–1949.
    1. Rochat A, Kobayashi K, Barrandon Y. Location of stem cells of human hair follicles by clonal analysis. Cell. 1994;76:1063–1073.
    1. Safer JD, Crawford TM, Holick MF. Topical thyroid hormone accelerates wound healing in mice. Endocrinology. 2005;146:4425–4430.
    1. Safer JD, Fraser LM, Ray S, Holick MF. Topical triiodothyronine stimulates epidermal proliferation, dermal thickening, and hair growth in mice and rats. Thyroid. 2001;11:717–724.
    1. Silva-Vargas V, Lo Celso C, Giangreco A, Ofstad T, Prowse DM, Braun KM, Watt FM. Beta-catenin and Hedgehog signal strength can specify number and location of hair follicles in adult epidermis without recruitment of bulge stem cells. Dev Cell. 2005;9:121–131.
    1. Sirakov M, Skah S, Lone IN, Nadjar J, Angelov D, Plateroti M. Multi-level interactions between the nuclear receptor TRalpha1 and the WNT effectors beta-catenin/Tcf4 in the intestinal epithelium. PLoS One. 2012;7:e34162.
    1. Slominski A, Wortsman J. Neuroendocrinology of the skin. Endocr Rev. 2000;21:457–487.
    1. Slominski A, Wortsman J, Kohn L, Ain KB, Venkataraman GM, Pisarchik A, Chung JH, Giuliani C, Thornton M, Slugocki G, Tobin DJ. Expression of hypothalamic-pituitary-thyroid axis related genes in the human skin. J Invest Dermatol. 2002;119:1449–1455.
    1. Snippert HJ, Haegebarth A, Kasper M, Jaks V, van Es JH, Barker N, van de Wetering M, van den Born M, Begthel H, Vries RG, et al. Lgr6 marks stem cells in the hair follicle that generate all cell lineages of the skin. Science. 2010;327:1385–1389.
    1. Sun G, Shi YB. Thyroid hormone regulation of adult intestinal stem cell development: mechanisms and evolutionary conservations. Int J Biol Sci. 2012;8:1217–1224.
    1. Taylor G, Lehrer MS, Jensen PJ, Sun TT, Lavker RM. Involvement of follicular stem cells in forming not only the follicle but also the epidermis. Cell. 2000;102:451–461.
    1. Tiede S, Bohm K, Meier N, Funk W, Paus R. Endocrine controls of primary adult human stem cell biology: thyroid hormones stimulate keratin 15 expression, apoptosis, and differentiation in human hair follicle epithelial stem cells in situ and in vitro. Eur J Cell Biol. 2010;89:769–777.
    1. Trempus CS, Morris RJ, Bortner CD, Cotsarelis G, Faircloth RS, Reece JM, Tennant RW. Enrichment for living murine keratinocytes from the hair follicle bulge with the cell surface marker CD34. J Invest Dermatol. 2003;120:501–511.
    1. van Beek N, Bodo E, Kromminga A, Gaspar E, Meyer K, Zmijewski MA, Slominski A, Wenzel BE, Paus R. Thyroid hormones directly alter human hair follicle functions: anagen prolongation and stimulation of both hair matrix keratinocyte proliferation and hair pigmentation. J Clin Endocrinol Metab. 2008;93:4381–4388.
    1. Vidal VP, Chaboissier MC, Lutzkendorf S, Cotsarelis G, Mill P, Hui CC, Ortonne N, Ortonne JP, Schedl A. Sox9 is essential for outer root sheath differentiation and the formation of the hair stem cell compartment. Curr Biol. 2005;15:1340–1351.
    1. Waikel RL, Kawachi Y, Waikel PA, Wang XJ, Roop DR. Deregulated expression of c-Myc depletes epidermal stem cells. Nat Genet. 2001;28:165–168.
    1. Watt FM, Frye M, Benitah SA. MYC in mammalian epidermis: how can an oncogene stimulate differentiation. Nat Rev Cancer. 2008;8:234–242.
    1. Yang L, Wang L, Yang X. Disruption of Smad4 in mouse epidermis leads to depletion of follicle stem cells. Mol Biol Cell. 2009;20:882–890.
    1. Zhang J, He XC, Tong WG, Johnson T, Wiedemann LM, Mishina Y, Feng JQ, Li L. Bone morphogenetic protein signaling inhibits hair follicle anagen induction by restricting epithelial stem/progenitor cell activation and expansion. Stem Cells. 2006;24:2826–2839.

Source: PubMed

3
Abonnieren