You are on page 1of 4

DOI: 10.1111/exd.

12024
www.blackwellpublishing.com/EXD
Viewpoint

Androgen actions on the human hair follicle: perspectives


Shigeki Inui and Satoshi Itami
Department of Regenerative Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan
Correspondence: Shigeki Inui, MD, PhD, Department of Regenerative Dermatology, Graduate School of Medicine, Osaka University,
2-2 Yamadaoka, Suita-shi, Osaka 565-0871, Japan, Tel.: +81-6-6879-3960, Fax: +81-6-6879-3962, e-mail: inui@r-derma.med.osaka-u.ac.jp

Abstract: Androgens stimulate beard growth but suppress hair DP cells and stem cell factor for positive regulation of hair growth
growth in androgenetic alopecia (AGA). This condition is known in beard and negative regulation of balding DP cells. Moreover,
as ‘androgen paradox’. Human pilosebaceous units possess AGA involves crosstalk between androgen and Wnt/b-catenin
enough enzymes to form the active androgens testosterone and signalling. Finally, recent data on susceptibility genes have
dihydrotestosterone. In hair follicles, 5a-reductase type 1 and 2, provided us with the impetus to investigate the molecular
androgen receptors (AR) and AR coactivators can regulate pathogenesis of AGA.
androgen sensitivity of dermal papillae (DP). To regulate hair
Key words: 5a-reductase – androgen – androgen receptor – growth factor
growth, androgens stimulate production of IGF-1 as positive
– Wnt
mediators from beard DP cells and of TGF-b1, TGF-b2, dickkopf1
and IL-6 as negative mediators from balding DP cells. In addition, Accepted for publication 31 August 2012
androgens enhance inducible nitric oxide synthase from occipital

Scope (FPHL) have not been sufficiently studied, the effect of androgen
Hair growth and cycling are regulated by many hormones (1), on FPHL has been merely speculated on through clinical observa-
and the effect of androgens, in particular, has been well known tion of antiandrogenic therapy. A controlled trial of oral cyproter-
for a long time. However, many questions about androgen metab- one acetate (CPA) for FPHL showed that it significantly increases
olism and function in hair follicles remain unanswered. In this the ratio of anagen to telogen in the fronto-cranial scalp (12).
review, we have examined published evidence and reconsidered However, Vexiau et al. (13) failed to show that CPA stimulates
the perspective on molecular physiology and pathophysiology of hair growth in women with FPHL, and moreover, FPHL can also
the effect of androgens on hair growth. occur in complete androgen insensitivity syndrome (14), speculat-
Clinical view of androgens and hair growth ing other possible pathogenetic factors besides an androgenetic
Androgens regulate human hair growth, and their effects vary aetiology (15). Observation of the positive relationship between
depending on body sites. Before puberty, there is only vellus hair hormones and axillary hair growth in 177 normal women has
in the pubic and axillary areas of males and females, but when suggested that weak adrenal androgens such as dehydroepiandros-
androgens increase in puberty, vellus hair follicles develop into terone (DHEA) and dehydroepiandrosterone sulfate (DHEA-S),
terminal ones, producing larger, curlier and darker hair shafts. In rather than testosterone, may play dominant roles in the preserva-
males, androgens stimulate beard growth but suppress hair growth tion of female axillary hair (16).
in androgenetic alopecia (AGA) and this reciprocal effect is known Androgen metabolism in human pilosebaceous unit
as the ‘androgen paradox’ (2,3). Historically, the first scientific The weak proandrogens DHEA and DHEA-S are mainly produced
description of the effect of androgens on AGA was published by from the adrenal cortex, while androstenedione is produced in
James B Hamilton in 1942 (4) based on his clinical observation of equal quantities from the adrenal cortex and ovaries, and in lesser
androgen induction of AGA in the males with testicular insuffi- quantity by the testes (17,18). These prohormones are then
ciency. On the other hand, Margaret Chieffi was the first to scien- converted into more potent androgens such as testosterone and
tifically prove the positive action of androgen on beard growth in dihydrotestosterone (DHT). Testosterone, a major circulating
males in clinical experiments to investigate the effect of testoster- androgen, is mainly produced by testes from puberty onward but
one injection on beard growth of elderly males (5). In male in reproductive-age females, it is secreted from the adrenal cortex
pseudohermaphrodites, with 5aR2 deficiency, normal axillary and and ovaries through conversion from androstenedione. DHT is
female-pattern pubic hair growth but no or little beard growth synthesized in peripheral tissues including skin. In addition,
and no AGA is seen (6–9), indicating that 5aR2 is necessary for DHEA, DHEA-S and androstenedione are converted by sebocytes,
beard growth and AGA development but not for pubic and axil- sweat glands and dermal papilla (DP) cells into more potent
lary hair growth. In female hirsutism patients, androgenic factors, androgens in the skin (19).
including polycystic ovary syndrome, are responsible for up to As the first step of androgen metabolism in human piloseba-
80% of the condition (10), suggesting that androgens can regulate ceous units, steroid sulfatase in DP (20) can hydrolyze DHEA-S to
hair growth in not only males but also in females, so that oral DHEA (Fig. 1). Next, 3b-hydroxysteroid dehydrogenase (3b-HSD)
antiandrogenic contraceptives are recommended as a first-line type 1 in sebaceous glands (21) and DP (22) converts DHEA into
treatment (11). Because the basic features of androgens in relation androstenedione. Furthermore, androstenedione is converted into
to female hair growth and female AGA or female pattern hair loss testosterone by 17b-hydroxysteroid dehydrogenase (17b-HSD),

ª 2012 John Wiley & Sons A/S


168 Experimental Dermatology, 2013, 22, 168–171
Androgen actions on the human hair follicle

DHEA-S Estrone (E1) expressed in all scalp sites (36). These discrepancies may be
ascribed to differences in sampling and methodology but a more
Sulfotransferases Steroid sulfatase Aromatase
plausible explanation has been provided by the report by Asada
3β-HSD 5α-reductase (37) that 5aR1 mRNA is expressed in all components of scalp hair
DHEA Androstenedione 5α-Androstenedione
follicles, but that 5aR2 mRNA is constitutively expressed only in
freshly microdissected scalp DP and connective tissue sheath. The
17β-HSD 17β-HSD 17β-HSD
expression of 5aR2 disappears during subcultivation, suggesting
3β-HSD 5α-reductase that 5aR2 is unstable in scalp cells in vitro. Finally, the clinical
Androstenediol Testosterone 5α-dihydrotestosterone phenotype of pseudohermaphrodites with 5aR2 deficiency as men-
Aromatase tioned above presents very strong evidence that beard and AGA
formation is dependent on 5aR2 activity but not 5aR1. As for
17β-Estradiol (E2) gender differences, 5aR1 and 5aR2 contents in female frontal hair
Figure 1. Androgen metabolism in skin. DHEA, dehydroepiandrosterone; DHEA-S,
follicles were 3 and 3.5 times less than in male frontal follicles
dehydroepiandrosterone sulfate; 3b-HSD, 3b-hydroxysteroid dehydrogenase; (27), respectively, indicating that female AGA/FPHL is less depen-
17b-HSD, 17b-hydroxysteroid dehydrogenase.
dent on 5a-reductase, which may explain the inefficiency of finas-
teride for FPHL (38).
whose enzyme activity has been detected in DP (23). Additionally, Androgen receptors
human sebaceous glands provide the cellular machinery for tran- The action of steroid hormones including androgens is mediated
scribing the genes for 17b-HSD types 1–5 (21,24,25), indicating by the intracellular nuclear receptors, which function as hormone-
the possible role of sebaceous glands for the regulation of local inducible transcription factors. Previously reported immunohisto-
androgen metabolism. Alternatively, DHEA can be converted into chemical results for distribution of androgen receptors (AR) in
androstenediol and testosterone by 17b-HSD and 3b-HSD in the hair follicles were inconsistent as summarized in Table S1 (39–
pilosebaceous unit, respectively and in that order (26). Cyto- 50). AR is generally expressed in the DP and sebaceous glands,
chrome P450 aromatase is required for bioconversion of andro- but the results for its expression in follicular epithelium are con-
gens to oestrogens. Aromatase activity is detectable in hair follicles troversial, probably due to differences in the antibodies used and
(27,28), and its expression in the outer root sheath of anagen hair in the skin portions of the hair follicles examined. More recent
follicles and in sebaceous glands (29), suggesting the presence of a data show that AR is not found in the outer root sheath, hair bulb
local balance system for androgens and oestrogens and that hair and bulge (45,48,49). In line with these results, endogenous AR
follicles function as oestrogen targets and sources (30). A compari- transactivity was undetectable in Hacat keratinocytes, indicating
son of aromatase content in frontal hair follicles from men and that human keratinocytes are unlikely to be target cells for andro-
women with pattern baldness has shown that it is six times greater gens (51). In addition, AR expression was detected in only the DP
in women (27), which has led to speculation that this difference of red deer hair follicles (47). However, it may be possible that
may account for the difference in clinical presentations of pattern AR is expressed in the hair epithelium of specific body sites or
baldness. hair diseases. Indeed, AR mRNA has been detected in the micro-
5a-reductase type 1 and 2 dissected inner and outer root sheath (37) of male and female
The two isoforms of human 5aR1 and 5aR2 are encoded by the sexual hairs, and it was found that type I hair keratin hHa7 is
Steroid5-alpha-reductase 1 and 2 (SRDA1 and SRDA2) genes, directly regulated by androgens through the androgen response
respectively. They convert testosterone to a more potent androgen element in its promoter (49).
DHT in target cells. 5aR1 consists of 259 amino acids and has an Previous semiquantitative RT-PCR as well as immunohisto-
optimal pH of 6–9, whereas 5aR2 consists of 254 amino acids and chemical and hormone binding assays demonstrated that AR
has an optimal pH of 5.5. 5aR1 is detected in various androgen- expression is significantly higher in beard and AGA DP than in
independent organs such as liver and brain, while 5aR2 is non-bald occipital scalp cells (36,50,52–55), indicating that AR is
predominantly observed in androgen-dependent organs such as one of the key molecules which regulate androgen sensitivity in
epididymis and prostate (31). In 1972, Takayasu and Adachi first DP. A recent study demonstrated that DNA methylation of the
identified 5a-reductase activity in human growing and resting hair AR promoter is increased in hair follicles from occipital scalp
follicles plucked from male scalps and found that the optimal pH compared with those from vertex AGA scalp, indicating efficacy
was 7.5, not 5.5 (32). Consistent with this finding, it was reported by the DNA methylation for protecting occipital hair follicles
that an inhibitor of 5aR1 and 5aR1/2 can suppress endogenous against AR expression (56). Moreover, AR content in female fron-
5a-reductase activity in plucked hairs from females but selective tal hair follicles was found to be approximately 40% lower than in
5aR2 inhibitors cannot (33). These observations seem to indicate male frontal follicles (27), again pointing to a possible cause of
that 5aR1 may be dominant in hair follicles. Another more recent the different presentations of pattern baldness.
study detected activity of both 5aR1 and 5aR2 in microdissected Downstream androgen receptors and hair growth
hair follicles and found that it was higher in balding hair follicles Downstream of AR there are many AR coregulators such as
than occipital hair follicles from both men and women (27). In coactivators, integrators or corepressors. One of the AR coactiva-
contrast with plucked hair follicles, beard DP cells show higher tors, Hic-5/ARA55 (57), is highly expressed in DP cells from
5aR2 activity than occipital DP cells in vitro (34,35). At the androgen-sensitive sites such as AGA and beard, suggesting
mRNA level, 5aR2 mRNA is higher in DP cells from AGA and that Hic-5/ARA55 can enhance androgen sensitivity in DP (58).
beard than in those from the occipital scalp while 5aR1 is equally On the other hand, another in situ labelling study showed that

ª 2012 John Wiley & Sons A/S


Experimental Dermatology, 2013, 22, 168–171 169
Inui and Itami

expression of another AR coactivator, ARA70, was weaker in the kinase-b in DP cells, but that Wnt signalling activation by LiCl
DP of balding recipient areas than in those from the donor areas restores the ability of DHT-treated DP cells to induce differentia-
(59), thus indicating that selective AR coactivators may be tion in the coculture of HFS cells and DP cells from AGA (75).
involved in the pathogenesis of AGA. These findings suggest that the crosstalk between Wnt/b-catenin
Embryonic induction of hair follicles and maintaining the func- and androgen receptor signalling performs an important function
tional integrity of adult follicles are governed by the interaction in androgen’s action on hair growth. Indeed, a recent phase 1 trial
between DP and follicular epithelium, possibly through paracrine revealed intradermal administration of a ‘Hair Stimulating
mediators (60). Such mediators can, therefore, also be involved in Complex’ containing Wnt and follistatin improved hair growth in
androgen regulation for hair growth. In a study using a coculture subjects with AGA (76). As HFS cells are preserved in AGA, these
system of outer root sheath cells and beard DP cells, IGF-1 was signalling interactions represent potential therapeutic targets in
first identified as a testosterone-inducible positive paracrine medi- AGA (77).
ator from beard DP cells (45) (Table S2). In addition, testosterone Perspectives
reportedly induced autocrine stimulatory factors from beard DP Very recently, the large-scale meta-analysis of seven genome-wide
cells (61), which suggests that autocrine behaviour is also involved association studies for early-onset AGA in 12 806 European indi-
in androgen regulation for beard growth. Furthermore, TGF-b1 viduals identified susceptibility loci at chromosomes 1, 2, 7, 17,
(62,63), TGF-b2 (64), dickkopf1 (65) and IL-6 (66) have been 18, 20 and X (78). Among the genes harbouring or nearest to the
identified as androgen-inducible negative mediators for AGA SNP, FOXA2 encodes a transcription factor Foxa2, which interacts
development in a number of studies using various in vitro experi- with AR through DNA biding domain to regulate gene expression
mental methods (Table S2), thus providing new clues for clarify- (79), and HDAC4 (histone deacetylase 4) encodes HDAC4, which
ing more details of androgen action in AGA. Moreover, the plays a critical role in suppression of AR transcriptional activity
finding that DHT increases inducible nitric oxide (NO) synthase through interacting with other transcriptional factors such as
(iNOS) from occipital DP cells suggests that iNOS and NO are androgen receptor corepressor-19 (80) and CR6-interacting factor
downstream effectors of AR in DP cells (67) (Table S2). Other 1 (81). From these findings, the regulation of AR transcriptional
reported findings are that beard DP cells produce more stem cell activity is critical for AGA development. In this line, AR coactiva-
factor (SCF) than non-balding scalp DP cells (68) and conversely tors/coreppressors and other pathways influencing AR activity can
that balding DP cells produce less SCF than non-balding scalp DP be hopeful targets to study for delineating AGA pathogenesis. On
cells (69). However, testosterone did not alter the amount of SCF the other hand, the recent microarray study using human scalp
from balding DP cells. These findings may link hair growth skin revealed that prostaglandin D2 synthase (PTGDS) is elevated
regulation to melanocyte activity because this factor stimulates at the mRNA and protein levels in bald scalp compared with
melanogenesis (70). haired scalp of men with AGA and the product of PTGDS enzyme
Crosstalk between androgen and Wnt/b-catenin activity, prostaglandin D2 is similarly elevated in bald scalp (82),
signalling suggesting that pathogenic mechanisms beyond androgen pathway
The Wnt signalling pathway is essential for maintaining the hair- play important roles in AGA. Alternatively, there may be any link-
inducing activity of DP cells (71) and for the development and age between AR and prostaglandin D2. We are still on the way to
regeneration of hair follicles in vivo (72,73). Recent reports have figure out the complex physiological and pathogenic network in
promoted the notion that crosstalk occurs between androgen and androgen regulation for hair growth and AGA pathogenesis.
Wnt/b-catenin signalling in AGA pathogenesis. DHT was shown Acknowledgement
to suppress Hacat keratinocyte proliferation stimulated by Wnt-3a We appreciate helpful discussion for this manuscript with Dr. Oh Sang
in a coculture of Hacat and DP cells from AGA (74). It was also Kwon at Department of Dermatology, Seoul National University College of
found that DHT suppresses Lef/Tcf-mediated transcriptional activ- Medicine. Writing of this viewpoint was supported in part by Grants-
ity in DP cells of AGA. However, these phenomena could not be in-Aid for Scientific Research (KAKENHI) to Sh I. Sh I wrote the study,
observed in DP cells of non-AGA males. Moreover, a very recent and Sa I revised it critically and approved the submitted and final version.
study showed that DHT inhibits hair follicle stem (HFS) cell Conflict of interests
differentiation, possibly through upregulation of glycogen synthase The authors have declared no conflicting interests.

References
1 Al-Nuaimi Y, Baier G, Watson R E et al. Exp Der- 11 Blume-Peytavi U, Hahn S. Dermatol Ther 2008: 19 Zouboulis C C. Horm Res 2000: 54: 230–242.
matol 2010: 19: 707–713. 21: 329–339. 20 Hoffmann R, Rot A, Niiyama S et al. J Invest
2 Randall V A. Semin Cell Dev Biol 2007: 18: 274–285. 12 Peereboom-Wynia J D, Van der Willigen A H, Dermatol 2001: 117: 1342–1348.
3 Randall V A, Hibberts N A, Thornton M J et al. Van Joost T et al. Acta Derm Venereol 1989: 21 Fritsch M, Orfanos C E, Zouboulis C C. J Invest
Horm Res 2000: 54: 243–250. 69: 395–398. Dermatol 2001: 116: 793–800.
4 Hamilton J. Am J Anat 1942: 71: 451–480. 13 Vexiau P, Chaspoux C, Boudou P et al. Br J Der- 22 Hoffmann R. Eur J Dermatol 2001: 11: 296
5 Chiieffi M. J Gerontol 1949: 4: 200–204. matol 2002: 146: 992–999. –300.
6 Imperato-McGinley J, Guerrero L, Gautier T 14 Cousen P, Messenger A. Br J Dermatol 2010: 23 Sato T, Tadokoro T, Sonoda T et al. J Dermatol
et al. Science 1974: 186: 1213–1215. 162: 1135–1137. Sci 1999: 19: 123–125.
7 Griffin J E, Wilson J D. N Engl J Med 1980: 302: 15 Langan E A, Paus R. Br J Dermatol 2010: 163: 24 Thiboutot D, Martin P, Volikos L et al. J Invest
198–209. 1140–1141. author reply 1141-1142. Dermatol 1998: 111: 390–395.
8 Andersson S, Berman D M, Jenkins E P et al. 16 Ishihara F, Komatsu M, Yamada T et al. Horm 25 Hoppe U, Holterhus P M, Wunsch L et al. J Mol
Nature 1991: 354: 159–161. Metab Res 1993: 25: 34–36. Med (Berl) 2006: 84: 651–659.
9 Wilson J D. Biol Reprod 1992: 46: 168–173. 17 Rosenfield R. Adv Pediatr 1972: 19: 171–213. 26 Takayasu S. Int J Dermatol 1979: 18: 681–692.
10 Blume-Peytavi U, Atkin S, Shapiro J et al. Eur J 18 Zouboulis C C, Chen W C, Thornton M J et al. 27 Sawaya M E, Price V H. J Invest Dermatol 1997:
Dermatol 2009: 19: 597–602. Horm Metab Res 2007: 39: 85–95. 109: 296–300.

ª 2012 John Wiley & Sons A/S


170 Experimental Dermatology, 2013, 22, 168–171
Androgen actions on the human hair follicle

28 Schweikert H U, Milewich L, Wilson J D. J Clin 48 Thornton M J, Taylor A H, Mulligan K et al. J In- 70 Besmer P, Manova K, Duttlinger R et al. Dev
Endocrinol Metab 1975: 40: 413–417. vestig Dermatol Symp Proc 2003: 8: 100–103. Suppl 1993: 125–137.
29 Sawaya M E, Penneys N S. J Cutan Pathol 1992: 49 Jave-Suarez L F, Langbein L, Winter H et al. J 71 Kishimoto J, Burgeson R E, Morgan B A. Genes
19: 309–314. Invest Dermatol 2004: 122: 555–564. Dev 2000: 14: 1181–1185.
30 Ohnemus U, Uenalan M, Inzunza J et al. Endocr 50 Kwon O S, Han J H, Yoo H G et al. J Dermatol 72 Andl T, Reddy S T, Gaddapara T et al. Dev Cell
Rev 2006: 27: 677–706. Sci 2004: 36: 176–179. 2002: 2: 643–653.
31 Russell D W, Berman D M, Bryant J T et al. 51 Inui S, Itami S, Pan H J et al. J Dermatol Sci 73 Ito M, Yang Z, Andl T et al. Nature 2007: 447:
Recent Prog Horm Res 1994: 49: 275–284. 2000: 23: 87–92. 316–320.
32 Takayasu S, Adachi K. J Clin Endocrinol Metab 52 Ando Y, Yamaguchi Y, Hamada K et al. Br J 74 Kitagawa T, Matsuda K, Inui S et al. J Clin Endo-
1972: 34: 1098–1101. Dermatol 1999: 141: 840–845. crinol Metab 2009: 94: 1288–1294.
33 Gerst C, Dalko M, Pichaud P et al. Exp Dermatol 53 Randall V A, Thornton M J, Hamada K et al. J 75 Leiros G J, Attorresi A I, Balana M E. Br J Der-
2002: 11: 52–58. Invest Dermatol 1992: 98: 86S–91S. matol 2012: 166: 1035–1042.
34 Itami S, Kurata S, Takayasu S. J Invest Dermatol 54 Randall V A, Thornton M J, Messenger A G. J 76 Zimber M P, Ziering C, Zeigler F et al. J Drugs
1990: 94: 150–152. Endocrinol 1992: 133: 141–147. Dermatol 2011: 10: 1308–1312.
35 Itami S, Kurata S, Sonoda T et al. J Invest Der- 55 Itami S, Kurata S, Sonoda T et al. Br J Dermatol 77 Garza L A, Yang C C, Zhao T et al. J Clin Invest
matol 1991: 96: 57–60. 1995: 132: 527–532. 2011: 121: 613–622.
36 Nakanishi S, Itami S, Adachi K et al. Expression 56 Cobb J E, Wong N C, Yip L W et al. Br J Derma- 78 Li R, Brockschmidt F F, Kiefer A K et al. PLoS
of androgen receptor, type I and type II 5a- tol 2011: 165: 210–213. Genet 2012: 8: e1002746.
reductase in human dermal papilla cells. In: Ne- 57 Fujimoto N, Yeh S, Kang H Y et al. J Biol Chem 79 Yu X, Gupta A, Wang Y et al. Ann N Y Acad
ste D, Randall V A, eds. Hair Research for the 1999: 274: 8316–8321. Sci 2005: 1061: 77–93.
Next Millennium. Amsterdam: Elsevier Publishers 58 Inui S, Fukuzato Y, Nakajima T et al. J Invest 80 Jeong B C, Hong C Y, Chattopadhyay S et al.
BV, 1996: 333–337. Dermatol 2007: 127: 2302–2306. Mol Endocrinol 2004: 18: 13–25.
37 Asada Y, Sonoda T, Ojiro M et al. J Clin Endocri- 59 Lee P, Zhu C C, Sadick N S et al. J Cutan Pathol 81 Suh J H, Shong M, Choi H S et al. Mol Endocri-
nol Metab 2001: 86: 2875–2880. 2005: 32: 567–571. nol 2008: 22: 33–46.
38 Price V H, Roberts J L, Hordinsky M et al. J Am 60 Oliver R F, Jahoda C A. Clin Dermatol 1988: 6: 82 Garza L A, Liu Y, Yang Z et al. Sci Transl Med
Acad Dermatol 2000: 43: 768–776. 74–82. 2012: 4: 126ra134.
39 Blauer M, Vaalasti A, Pauli S L et al. J Invest Der- 61 Thornton M J, Hamada K, Messenger A G et al. Supporting Information
matol 1991: 97: 264–268. J Invest Dermatol 1998: 111: 727–732.
40 Ruizeveld de Winter J A, Trapman J, Vermey M 62 Inui S, Fukuzato Y, Nakajima T et al. J Investig Additional Supporting Information may be found in
et al. J Histochem Cytochem 1991: 39: 927–936. Dermatol Symp Proc 2003: 8: 69–71. the online version of this article:
41 Choudhry R, Hodgins M B, Van der Kwast T H 63 Inui S, Fukuzato Y, Nakajima T et al. Faseb J Data S1. Impetus from genetic studies of AGA.
et al. J Endocrinol 1992: 133: 467–475. 2002: 16: 1967–1969. Table S1. Summary of immunohistochemical studies
42 Kimura N, Mizokami A, Oonuma T et al. J Histo- 64 Hibino T, Nishiyama T. J Dermatol Sci 2004: 35: of androgen receptor localization in pilosebaceous unit.
chem Cytochem 1993: 41: 671–678. 9–18. Table S2. Summary of cell culture studies of andro-
43 Liang T, Hoyer S, Yu R et al. J Invest Dermatol 65 Kwack M H, Sung Y K, Chung E J et al. J Invest gen-inducible factors in dermal papilla cells.
1993: 100: 663–666. Dermatol 2008: 128: 262–269. Please note: Wiley-Blackwell are not responsible for
44 Iwamura M, Abrahamsson P A, Benning C M 66 Kwack M H, Ahn J S, Kim M K et al. J Invest the content or functionality of any supporting materials
et al. J Histochem Cytochem 1994: 42: 783–788. Dermatol 2012: 132: 43–49. supplied by the authors. Any queries (other than miss-
45 Itami S, Kurata S, Takayasu S. Biochem Biophys 67 Wolf R, Schonfelder G, Paul M et al. J Mol Med ing material) should be directed to the corresponding
Res Commun 1995: 212: 988–994. (Berl) 2003: 81: 110–117. author for the article.
46 Bird J, Li X, Lei Z M et al. J Clin Endocrinol 68 Hibberts N A, Messenger A G, Randall V A. Bio-
Metab 1998: 83: 1776–1782. chem Biophys Res Commun 1996: 222: 401–405.
47 Thornton M J, Hibberts N A, Street T et al. J 69 Randall V A, Jenner T J, Hibberts N A et al. J
Endocrinol 2001: 168: 401–408. Endocrinol 2008: 197: 11–23.

ª 2012 John Wiley & Sons A/S


Experimental Dermatology, 2013, 22, 168–171 171

You might also like