10
Endogenous Retinoids in the Pathogenesis of Alopecia Areata F. Jason Duncan 1,7 , Kathleen A. Silva 2,7 , Charles J. Johnson 1,7 , Benjamin L. King 2 , Jin P. Szatkiewicz 2 , Sonya P. Kamdar 2 , David E. Ong 3 , Joseph L. Napoli 4 , Jinshan Wang 4 , Lloyd E. King Jr 3 , David A. Whiting 5 , Kevin J. McElwee 6 , John P. Sundberg 2,3 and Helen B. Everts 1 Alopecia areata (AA) is an autoimmune disease that attacks anagen hair follicles. Gene array in graft-induced C3H/ HeJ mice revealed that genes involved in retinoic acid (RA) synthesis were increased, whereas RA degradation genes were decreased in AA compared with sham controls. This was confirmed by immunohistochemistry in biopsies from patients with AA and both mouse and rat AA models. RA levels were also increased in C3H/HeJ mice with AA. C3H/HeJ mice were fed a purified diet containing one of the four levels of dietary vitamin A or an unpurified diet 2 weeks before grafting and disease progression followed. High vitamin A accelerated AA, whereas mice that were not fed vitamin A had more severe disease by the end of the study. More hair follicles were in anagen in mice fed high vitamin A. Both the number and localization of granzyme B–positive cells were altered by vitamin A. IFNg was also the lowest and IL13 highest in mice fed high vitamin A. Other cytokines were reduced and chemokines increased as the disease progressed, but no additional effects of vitamin A were seen. Combined, these results suggest that vitamin A regulates both the hair cycle and immune response to alter the progression of AA. Journal of Investigative Dermatology (2013) 133, 334–343; doi:10.1038/jid.2012.344; published online 27 September 2012 INTRODUCTION Alopecia areata (AA) is an autoimmune, nonscarring, hair loss disease affecting up to 1.7% of humans (Safavi et al., 1995). Current treatments are often ineffective in inducing prolonged remission (Tosti and Duque-Estrada, 2009; Harries et al., 2010). AA is characterized by a loss of hair follicle immune privilege, increased IFN gamma (IFNg) and other T helper 1 (Th1) cytokines, and an increase in CD8 þ T cells (McElwee et al., 1996; Gilhar et al., 2005; King et al., 2008). NK or NKT cells and T regulatory (Tregs) cells may also be involved (McElwee et al., 2005; Ito et al., 2008; Petukhova et al., 2010). Studies suggest that AA is a complex polygenetic disease (Sundberg et al., 2004; Petukhova et al., 2010). Little is known about the environmental factors, such as diet, that impact the course of AA. Several interactions between retinoids and immunity exist (Duriancik et al., 2010). Vitamin A deficiency impairs the development of cell-mediated immunity (Smith et al., 1987) and promotes Th1 responses while delaying Th2 development (Cantorna et al., 1994). Recently, additional T-cell subtypes were appreciated as important in autoimmunity, including Th17 and T Tregs cells, which are both regulated by vitamin A to maintain gut immune tolerance (Stockinger et al., 2007; Sojka et al., 2008). Retinoic acid (RA) synthesis occurs in dendritic cells (DCs) in the gut (Iwata et al., 2004), which increases FOXP3-positive Tregs (Coombes et al., 2007) and inhibits Th17 cell development (Mucida et al., 2007) via RA receptor alpha (RARA) in vitro (Schambach et al., 2007). Inducing in vivo endogenous RA synthesis through activation of toll-like receptor 2 (Manicassamy et al., 2009) or PPARG agonist (Housley et al., 2009) inhibited Th17 cells and increased FOXP3. Exogenous RA inhibited Th17 cells but had no effect on FOXP3 (Xiao et al., 2008), suggesting that RA needs to be induced in a precise location endogenously to maintain gut immune tolerance. Similar to the gut, skin has a major barrier function. DCs in the dermis have similar characteristics to those in the gut, including langerin expression (Bursch et al., 2007). Mouse ear dermal DCs had aldehyde dehydrogenase activity and induced FOXP3-positive See related commentary on pg 285 ORIGINAL ARTICLE 1 Department of Nutrition, The Ohio State University, Columbus, Ohio, USA; 2 The Jackson Laboratory, Bar Harbor, Maine, USA; 3 Vanderbilt University Medical Center, Nashville, Tennessee, USA; 4 University of California, Berkeley, Berkeley, California, USA; 5 Baylor Hair Research and Treatment Center, Dallas, Texas, USA and 6 University of British Columbia, Vancouver, British Columbia, Canada Correspondence: Helen B. Everts, Department of Nutrition, The Ohio State University, Columbus, Ohio 43210, USA. E-mail: [email protected] 7 These authors contributed equally to this work. Received 7 February 2012; revised 11 July 2012; accepted 2 August 2012; published online 27 September 2012 Abbreviations: AA, alopecia areata; ALDH1A1, 2, 3, retinal dehydrogenase1, 2, 3; CRABP2, cellular retinoic acid–binding protein II; CRABP2, cellular retinoic acid–binding protein II; DGAT1, diacylglycerol acyltransferase 1; DHRS9, dehydrogenase reductase SDR family member 9; GZMB, granzyme B; LRAT, lecithin:retinol acyltransferase; NKG2D, natural killer group 2D; RA, retinoic acid; Raldhs, retinal dehydrogenases; RARA, B, G, retinoic acid receptor alpha, beta, gamma; RBP1 (formerly CRBP), cellular retinol–binding protein 1; Roldhs, retinol dehydrogenases; STRA6, stimulated by retinoic acid 6; Th1, T helper 1; VAA, vitamin A adequate; VAD, vitamin A deficient; VAE, vitamin A excess; VAH, vitamin A high. Genes and RNA message expression are italicized, whereas proteins are not. Proteins from mice, rats, and humans are all capital letters. 334 Journal of Investigative Dermatology (2013), Volume 133 & 2013 The Society for Investigative Dermatology

Endogenous Retinoids in the Pathogenesis of Alopecia Areata · 2017. 2. 1. · Alopecia areata (AA) is an autoimmune, nonscarring, hair loss disease affecting up to 1.7% of humans

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

  • Endogenous Retinoids in the Pathogenesis ofAlopecia AreataF. Jason Duncan1,7, Kathleen A. Silva2,7, Charles J. Johnson1,7, Benjamin L. King2, Jin P. Szatkiewicz2,Sonya P. Kamdar2, David E. Ong3, Joseph L. Napoli4, Jinshan Wang4, Lloyd E. King Jr3, David A. Whiting5,Kevin J. McElwee6, John P. Sundberg2,3 and Helen B. Everts1

    Alopecia areata (AA) is an autoimmune disease that attacks anagen hair follicles. Gene array in graft-induced C3H/HeJ mice revealed that genes involved in retinoic acid (RA) synthesis were increased, whereas RA degradation geneswere decreased in AA compared with sham controls. This was confirmed by immunohistochemistry in biopsiesfrom patients with AA and both mouse and rat AA models. RA levels were also increased in C3H/HeJ mice with AA.C3H/HeJ mice were fed a purified diet containing one of the four levels of dietary vitamin A or an unpurified diet2 weeks before grafting and disease progression followed. High vitamin A accelerated AA, whereas mice that werenot fed vitamin A had more severe disease by the end of the study. More hair follicles were in anagen in mice fedhigh vitamin A. Both the number and localization of granzyme B–positive cells were altered by vitamin A. IFNg wasalso the lowest and IL13 highest in mice fed high vitamin A. Other cytokines were reduced and chemokinesincreased as the disease progressed, but no additional effects of vitamin A were seen. Combined, these resultssuggest that vitamin A regulates both the hair cycle and immune response to alter the progression of AA.

    Journal of Investigative Dermatology (2013) 133, 334–343; doi:10.1038/jid.2012.344; published online 27 September 2012

    INTRODUCTIONAlopecia areata (AA) is an autoimmune, nonscarring, hair lossdisease affecting up to 1.7% of humans (Safavi et al., 1995).Current treatments are often ineffective in inducing prolongedremission (Tosti and Duque-Estrada, 2009; Harries et al.,2010). AA is characterized by a loss of hair follicle immuneprivilege, increased IFN gamma (IFNg) and other T helper 1(Th1) cytokines, and an increase in CD8þ T cells (McElweeet al., 1996; Gilhar et al., 2005; King et al., 2008). NK or NKT

    cells and T regulatory (Tregs) cells may also be involved(McElwee et al., 2005; Ito et al., 2008; Petukhova et al., 2010).Studies suggest that AA is a complex polygenetic disease(Sundberg et al., 2004; Petukhova et al., 2010). Little is knownabout the environmental factors, such as diet, that impact thecourse of AA.

    Several interactions between retinoids and immunity exist(Duriancik et al., 2010). Vitamin A deficiency impairs thedevelopment of cell-mediated immunity (Smith et al., 1987)and promotes Th1 responses while delaying Th2 development(Cantorna et al., 1994). Recently, additional T-cell subtypeswere appreciated as important in autoimmunity, includingTh17 and T Tregs cells, which are both regulated by vitamin Ato maintain gut immune tolerance (Stockinger et al., 2007;Sojka et al., 2008). Retinoic acid (RA) synthesis occurs indendritic cells (DCs) in the gut (Iwata et al., 2004), whichincreases FOXP3-positive Tregs (Coombes et al., 2007) andinhibits Th17 cell development (Mucida et al., 2007) via RAreceptor alpha (RARA) in vitro (Schambach et al., 2007).Inducing in vivo endogenous RA synthesis through activationof toll-like receptor 2 (Manicassamy et al., 2009) or PPARGagonist (Housley et al., 2009) inhibited Th17 cells andincreased FOXP3. Exogenous RA inhibited Th17 cells buthad no effect on FOXP3 (Xiao et al., 2008), suggesting that RAneeds to be induced in a precise location endogenously tomaintain gut immune tolerance. Similar to the gut, skin hasa major barrier function. DCs in the dermis have similarcharacteristics to those in the gut, including langerinexpression (Bursch et al., 2007). Mouse ear dermal DCs hadaldehyde dehydrogenase activity and induced FOXP3-positive

    See related commentary on pg 285ORIGINAL ARTICLE

    1Department of Nutrition, The Ohio State University, Columbus, Ohio, USA;2The Jackson Laboratory, Bar Harbor, Maine, USA; 3Vanderbilt UniversityMedical Center, Nashville, Tennessee, USA; 4University of California,Berkeley, Berkeley, California, USA; 5Baylor Hair Research and TreatmentCenter, Dallas, Texas, USA and 6University of British Columbia, Vancouver,British Columbia, Canada

    Correspondence: Helen B. Everts, Department of Nutrition, The Ohio StateUniversity, Columbus, Ohio 43210, USA. E-mail: [email protected]

    7These authors contributed equally to this work.

    Received 7 February 2012; revised 11 July 2012; accepted 2 August 2012;published online 27 September 2012

    Abbreviations: AA, alopecia areata; ALDH1A1, 2, 3, retinal dehydrogenase1,2, 3; CRABP2, cellular retinoic acid–binding protein II; CRABP2, cellularretinoic acid–binding protein II; DGAT1, diacylglycerol acyltransferase 1;DHRS9, dehydrogenase reductase SDR family member 9; GZMB, granzyme B;LRAT, lecithin:retinol acyltransferase; NKG2D, natural killer group 2D; RA,retinoic acid; Raldhs, retinal dehydrogenases; RARA, B, G, retinoic acidreceptor alpha, beta, gamma; RBP1 (formerly CRBP), cellular retinol–bindingprotein 1; Roldhs, retinol dehydrogenases; STRA6, stimulated by retinoic acid6; Th1, T helper 1; VAA, vitamin A adequate; VAD, vitamin A deficient; VAE,vitamin A excess; VAH, vitamin A high. Genes and RNA message expressionare italicized, whereas proteins are not. Proteins from mice, rats, and humansare all capital letters.

    334 Journal of Investigative Dermatology (2013), Volume 133 & 2013 The Society for Investigative Dermatology

    http://dx.doi.org/10.1038/jid.2012.344mailto:[email protected]

  • Tregs in an RAR-dependent manner, although it was notconfirmed that these cells also expressed langerin or retinaldehydrogenase 2 (ALDH1A2, (Guilliams et al., 2010). Collec-tively, the results from these studies suggest that RA synthesiswithin the gut and skin DCs regulates immune tolerance.

    Excess RA leads to alopecia (Ruzicka et al., 1992; Ries andHess, 1999; Shih et al., 2009), which may result from manyfactors including dysregulated immune function. Vitamin Adeficiency leads to follicular hyperkeratosis and rupture inhumans and rodents (Wolbach and Howe, 1925; Girard et al.,2006). In rodents, vitamin A deficiency also leads to a thin haircoat that is frequently seen but rarely reported (Anzano et al.,1979, unpublished observation; Everts and Berdanier, 2002).The results from these studies suggest that precise RA levelsare needed for optimal hair follicle function.

    RA synthesis occurs locally in or near the cells where it willultimately be used. Precise spatial and temporal levels of RAin the skin are achieved by regulating several key steps incellular vitamin A metabolism: storage as retinyl esters, RAsynthesis, and RA degradation (Everts, 2012). In brief, vitaminA circulates as retinol bound to retinol-binding protein (RBP4).Retinol is transported into the cell via stimulation by RA 6(STRA6) and binds cellular retinol binding protein 1 (RBP1,aka CRBP). This bound retinol can either be esterified bylecithin:retinol acyltransferase (LRAT) for storage or reversiblyoxidized to retinal via retinol dehydrogenases, such asdehydrogenase reductase SDR family member 9 (DHRS9) orRDH10. Retinal is further oxidized to RA by retinaldehydrogenases 1–3 (ALDH1A1–3). RA is then sent to thenucleus with the assistance of cellular RA binding protein 2(CRABP2) to bind its RA receptors alpha, beta, and gamma(RARA, B, C) and activate the transcription of 500þ genes(Balmer and Blomhoff, 2002). When RBP1/CRBP is saturatedor absent, retinol can be esterified by acyl CoA: diacylglycerolacyltransferase 1 (DGAT1) or cleared by conversion to retinalby alcohol dehydrogenases 1–4. Retinal is then oxidized to RAvia ALDH1A1 and further metabolized by cytochrome P45026 family members (CYP26A1, B1, C1) with the assistance ofcellular RA binding protein 1 (CRABP1).

    To better understand AA, transcriptome analysis of C3H/HeJmice with AA was performed. As retinoid metabolism was notpart of the network analysis software, the expression ofretinoid metabolism genes was examined with the hypothesisthat RA metabolism was not altered in AA. Transcripts codingfor proteins metabolizing retinoids were altered, which wasconfirmed in biopsies from AA patients and rodent AA models.High dietary vitamin A accelerated disease progression andnumbers of hair follicles in anagen. Lack of vitamin A resultedin a more severe disease. A few immune factors were alsoaltered by diet, suggesting that retinoids alter AA by regulatingboth the hair cycle and immune response.

    RESULTSThe capacity for RA synthesis was increased in AAAnalysis of graft-induced AA transcripts revealed that theexpression of most genes involved in RA synthesiswas significantly increased (Figure 1a, Supplementary TablesS3–S6 online), whereas the expression of Rbp4 and RA

    degradation genes was significantly decreased (Figure 1b) inAA compared with sham controls at 10, 15, and sometimes 20weeks after grafting. Only Rbp1 (Crbp1), Crabp2, and Aldh1a3transcripts were significantly increased and Stra6 significantlydecreased in mice with spontaneous AA compared with wild-type C3H/HeJ mice (Figure 1c).

    Because differences in the hair cycle between the AA miceand controls were found (Supplementary Figure S2 online) andRA synthesis components changed during the hair cycle(Everts et al., 2007), these gene array results were confirmedusing immunohistochemistry (IHC) on human, DEBR rat, andC3H/HeJ mouse skin, with or without AA, using antibodiesagainst specific RA synthesis and degradation proteins tobetter control for hair cycle changes. RBP1/CRBP had thegreatest increase in immunoreactivity in biopsies of patientswith AA and both rodent models (Figure 2a–d andSupplementary Figure S3a and b online). RBP1/CRBP wasalso high in CBA/CaHN-Btkxnl/J, SWR/J, and A/J mice with AA,but not in C3H/HeOuJ mice with AA (SupplementaryFigure S4 online). DHRS9 and CRABP2 were also increasedin mice but only slightly increased in AA patients and DEBRrats (Figure 2e–h, q–t and Supplementary Figure S3c, d, g, andh online). ALDH1A1 and ALDH1A2 were increased inbiopsies from humans, but were not different in rodent models(Figure 2i–l and Supplementary Figure S6a–d online).ALDH1A3 was greatly increased in mice, absent from thepre-medulla in DEBR rats, and not different in biopsies fromhumans, although no pre-medulla was present in humansamples (Figure 2m–p and Supplementary Figure S3e,f online). Immunoreactivity of CYP26B1 was not differentbetween C3H/HeJ mice with AA or controls, but localizationchanged during the hair cycle (data not shown). DHRS9,ALDH1A1, and ALDH1A2 also localized to infiltratingimmune cells in biopsies from human patients with AA andC3H/HeJ mice (Figure 2f, h, j, l and Supplementary FigureS6a–d arrow online). In DEBR rats, immune cells expressedDHRS9 but not ALDH1A1 or ALDH1A2 (SupplementaryFigure S3d online, data not shown). ALDH1A2 colocalizedwith DC markers langerin and natural killer group 2D(NKG2D; Supplementary Figure S6e, f arrow online).

    Retinoid levels were measured by liquid chromatography/mass spectometry (LC/MS/MS) and HPLC to confirm thisexpression pattern. RA levels were significantly greater(Po0.05), whereas retinol levels were lower (P¼ 0.061), inAA mice compared with controls (Supplementary Figure S7aand b online). There was no difference in retinyl ester levels(Supplementary Figure S7c online).

    Dietary vitamin A altered the progression of AA

    To determine whether dietary vitamin A altered AA, C3H/HeJ mice were fed one of the five diets, starting 2 weeksbefore receiving a graft from a mouse with AA, and theprogression of the disease was analyzed. Ventral hair losswas significantly increased at 13, 14, and 18 weeks postgrafting in mice fed high vitamin A (12 IU, VAH) comparedwith mice fed unpurified chow (control, Figure 3a). Therewas also a trend at 14 weeks with mice fed VAH diet havingmore hair loss than mice fed the vitamin A–deficient diet

    FJ Duncan et al.Retinoid Metabolism in Alopecia Areata

    www.jidonline.org 335

    http://www.jidonline.org

  • (VAD, P¼0.055). Fifteen weeks after grafting, mice fed VAHdiet had significantly more epidermal hyperplasia in ventralskin compared with mice fed VAD or adequate vitamin A (4IU, VAA), or control diets (Figure 3b). Mice fed excessvitamin A (28 IU, VAE) also had significantly more epider-mal hyperplasia in ventral skin than mice fed VAD diet. Thepercentage of hair follicles in mid-anagen through mid-catagen, when active disease is seen, was also greater inmice fed VAH and VAE diets compared with the control dietin both ventral and dorsal skin (Figure 3c). In the dorsal skin,these findings were also significantly greater than mice fedVAA diet. By contrast, at 20 weeks post grafting, mice fedVAD diet had significantly more lymphocytes and outer rootsheath hyperplasia in ventral skin compared with all otherdiets (Figure 3d and e). Follicular dystrophy was significantlygreater in mice fed VAD, VAA, and VAH diets comparedwith mice fed control diet (Figure 3f). Follicular dystrophywas also significantly greater in mice fed VAD comparedwith mice fed VAE diets. Similar results were seen in dorsal

    skin, with mice fed VAD diet having significantly morefollicular dystrophy than mice fed VAH and VAE diets(Po0.05, data not shown). Mice fed VAA diet also hadmore follicular dystrophy than mice fed VAH diet (Po0.05,data not shown).

    There were no differences in body weight between any ofthe groups (Supplementary Figure S8a online). Mice fed thecontrol diet ate more food than any of the other diets(Po0.001), whereas mice fed VAH diet ate significantly lessthan mice fed VAD, VAE, and control diets (Po0.05;Supplementary Figure S8b online). Both food intake and bodyweight increased with time as expected, yet neither wassignificantly different between the mice receiving the AAand sham grafts (Supplementary Figure S8a and b online).

    Dietary vitamin A altered the number and localization ofgranzyme B (GZMB)–positive cells

    To determine how dietary vitamin A altered activated immunecells in AA, IHC of GZMB was performed and positive cells

    8

    –3

    –2

    –1

    0

    1

    6

    Fol

    d ch

    ange

    (A

    A v

    s. s

    ham

    )

    Fol

    d ch

    ange

    (A

    A v

    s. c

    ontr

    ol)

    Fol

    d ch

    ange

    (A

    A v

    s. s

    ham

    )

    4

    ** **

    ** ***

    *

    *

    Crbp1

    Crabp1Cyp26b1Dgat1Aldh1a1Adh1Rbp4Crabp2

    Rara

    Crbp2

    Aldh1a3

    Lrat

    0 5 10 15

    Weeks after grafting

    20 25 0 5 10

    Crb

    p1

    Crb

    p2

    Cra

    bp2

    Cyp

    26b1

    Cra

    bp1

    Lrat

    Ald

    h1a3

    15

    Weeks after grafting

    20 25

    2

    0

    4

    –4

    –2

    2

    **

    *0

    Rar

    a

    Dga

    t1A

    ldh1

    a1A

    dh1

    Rbp

    4S

    tra6

    Rbp4-Retinol

    Stra6

    Dgat1

    Adh1 Rdh1,11Rdh10Dhrs9#

    Retinylesters

    Lrat

    STORAGE

    Aldh1a1

    Crabp1- Retinoic acid

    Cyp26a1

    Crbp (Rbp1,2)-Retinal

    Crbp (Rbp1,2)-Retinal

    Aldh1a3

    Rara

    Crabp2- Retinoic acid

    RargRarb

    Aldh1a2

    Cyp26b1

    Aldh1a1

    Retinol

    Retinal

    DEGRADATION

    #Dhrs9 was not on microarray chip

    GENE ACTIVATION

    Reduced Increased

    Retinyl esters

    STORAGE

    Figure 1. Retinoid metabolism is altered in alopecia areata. Fold changes (FC) in (a) retinoic acid (RA) synthesis and (b) retinol degradation proteins determined

    by microarray analysis between C3H/HeJ mice grafted with alopecia areata skin and sham controls 5, 10, 15, and 20 weeks after surgery, or (c) between

    mice with spontaneous disease and unaffected mice. *Po0.05, qo0.05 all genes, **Po0.05, qo0.05 all genes except Lrat, Rara, and Cyp26b1, ***Po0.05,qo0.05 only Rbp4, Adh1, and Dgat1. (d) Diagram of retinoid metabolism with genes significantly altered highlighted. Red, FC4þ 3; pink, FC 0 to þ3;dark green, FCo� 2; light green, FC 0 to �2. AA, alopecia areata.

    FJ Duncan et al.Retinoid Metabolism in Alopecia Areata

    336 Journal of Investigative Dermatology (2013), Volume 133

  • counted in the bulb, superbular, isthmus, and infundibulum ofstaged hair follicles. GZMB was significantly greater in AAmice versus shams (Figure 4a, c, e and g). Further statisticalanalysis was performed with only AA mice with hair folliclesin mid-anagen through mid-catagen, when disease is seen.GZMB was greatest in mice fed VAE diet 15 weeks postgrafting in and around the isthmus (Figure 4d). GZMB peakedat 10 weeks in and around the isthmus for mice fed VAH diet,

    yet was high at both 10 and 15 weeks for mice fed VAA andcontrol diets. In contrast, mice fed VAD diet had a differentlocalization pattern with significantly more positive cells inand around the bulb at 10 and 15 weeks than mice fed theother diets (Figure 4h). At 10 weeks, most of the positive cellswere in and around the bulb and superbulbar, but by week 15these mice had higher numbers of cells in and around theisthmus (Figure 4d, f and h).

    Figure 2. Retinoic acid synthesis enzymes and binding proteins are increased in alopecia areata. Immunohistochemistry (IHC) was performed on (a, e, i, m, q)

    C3H/HeJ mouse skin collected 10 weeks after grafting with sham controls, (b, f, j, n, r) alopecia areata, (c, g, k, o, s) biopsies from human patients with tinea

    capitis controls, (d, h, l, p, t) or alopecia areata with antibodies against (a–d) cellular retinol–binding protein (CRBP/RBP)1, (e–h) dehydrogenase reductase

    SDR family member 9 (DHRS9), (i–l) 2, 3, retinal dehydrogenase 1, 2, 3 (ALDH1A1), (m–p) ALDH1A3, (q–t) and cellular retinoic acid–binding protein II

    (CRABP2). Bar¼ 50mm. Arrow, positive immune cells.

    FJ Duncan et al.Retinoid Metabolism in Alopecia Areata

    www.jidonline.org 337

    http://www.jidonline.org

  • High vitamin A increased IL13 and reduced IFNcCytokines are differentially regulated in AA (Carroll et al.,2002; Deeths et al., 2006; Freyschmidt-Paul et al., 2006;McPhee et al., 2012). ELISAs were performed to determinewhether Th1, Th2, or Th17 cytokine protein levels weredifferentially regulated by diet in mice. There were a fewdiet differences. Mice fed VAH diet had significantly moreIL13 than control chow-fed mice, but there were nodifferences between the AA and sham controls (Figure 5a).Levels of IFNg varied at different times with different diets,with low IFNg seen in mice fed VAH diet at 15 weeks andmice fed VAE diet at 20 weeks (Figure 5b).

    Cytokine production and PDL1 was reduced, whereas CXCL9and CCL5 was increased, in AA

    Protein levels of IL4, IL10, IL22, and IFNg were elevated in AAmice compared with sham mice at 5 weeks, but this was onlysignificant for IL4 (Supplementary Figure S9a–c and f online).

    All cytokines progressively decreased in the AA mice andincreased in the sham mice with time after grafting. By 20weeks post grafting, IL10, IFNg, IL17, IL21, and IL22 weresignificantly decreased in mice that received the AA graftcompared with sham controls (Supplementary Figure S9a–fonline). Two factors essential for tolerance, CTLA4 and PDL1(CD274), were also tested in the skin by ELISA and IHC,repectively. CTLA4 was significantly lower by 20 weeks, butnot significantly different between AA and sham mice(Supplementary Figure S9g online). In contrast, PDL1(CD274) was significantly lower in mice that received theAA graft versus sham graft 15 and 20 weeks post grafting(Supplementary Figure S9h online).

    Quantitative real-time PCR analysis revealed that IFNgmRNA levels were significantly increased in AA mice 15weeks post grafting compared with sham controls, althoughno diet effect was seen (Supplementary Figure S10 online).

    To better select additional immune factors to analyze, anELISArray (Qiagen, Frederick, MD) was performed on the basisof results from a gene microarray and quantitative real-timePCR arrays (McPhee et al., 2012, ELISArray data not shown).CCL5 (RANTES) and CXCL9 (MIG) were significantlyincreased in the skin of mice receiving AA grafts comparedwith sham controls at 10, 15, and 20 weeks post grafting(Supplementary Figure S11a and b online). Neither chemokinewas altered by dietary vitamin A. Data for the control-fed micewere previously published (McPhee et al., 2012).

    DISCUSSIONThis report shows that the expression of retinoid synthesisenzymes and binding proteins is increased in human patientswith AA and in two rodent models. DCs and NKG2D-positivecells also contained RA synthesis enzymes. RA levels werealso increased in C3H/HeJ mice with AA. Feeding high levelsof dietary vitamin A combined with increased RA synthesisaccelerated the onset of AA. Yet, not feeding vitamin Aresulted in more severe disease by the end of the study. Thisduality of responses suggests that precise vitamin A levels areneeded. Analysis of immune factors showed that dietaryvitamin A altered GZMB, IL13, and IFNg, and providedadditional findings on AA pathogenesis. Regulation of the haircycle may have also contributed to the accelerated disease.

    This report shows increased RA synthesis in AA. To ourknowledge, any studies of RA alterations in AA are previouslyunreported. RA levels were also increased in the skin-specific(Krt14-cre recombinase) Scd1tm2Ntam-null mice with cicatricialalopecia (Flowers et al., 2011). The expression of RA synthesiscomponents was also increased in patients and C57BL/6Jmice with cicatricial alopecia (Everts et al., in press). Thus,RA synthesis may be secondary to these diseases. Regardlessof how changes occurred, alteration in retinoid metabolismmay make patients with AA more sensitive to exogenousretinoids.

    Vitamin A toxicity leads to alopecia (Ruzicka et al., 1992;Ries and Hess, 1999; Shih et al., 2009). In mice, excess retinoland all transRA (Dgat1tm2Far null mice) within the basalepidermis and outer root sheath (Krt14 cre) led toprogressive cyclical alopecia with accelerated telogen to

    300

    250

    5 10 14

    0 IU4 IU12 IU28 IU

    * *

    Chow

    0 IU4 IU12 IU

    AB AB ABA

    A A

    B BB B

    28 IUChow

    18

    200

    150

    100

    100

    80

    A

    0 4 12 28 Chow

    Dorsal Ventral

    Per

    cent

    of h

    air

    folli

    cles

    in a

    nage

    n

    0 4 12 28 Chow

    Diet

    Diet

    0 4 12 28 ChowDiet

    0 4 12 28 ChowDiet

    AC

    A B B B B A B B B B

    B BC AC01234

    01234

    01234

    01234

    60

    40

    % E

    pide

    rmal

    hype

    rpla

    sia

    scor

    e

    % O

    RS

    hype

    rpla

    sia

    scor

    e

    % L

    ymph

    ocyt

    essc

    ore

    % D

    ystr

    ophy

    sco

    re

    20

    Hai

    r

    50

    0

    150

    100

    50

    00

    100

    80

    60

    40

    20

    0

    100

    80

    60

    40

    20

    0

    100 A AB AB BC C

    80

    60

    40

    20

    0

    Figure 3. Progression of alopecia areata (AA) was altered by dietary vitamin A.

    Mice were fed purified diets containing 0, 4, 12, or 28 IU dietary vitamin A/g

    diet, or a control chow diet, starting 2 weeks before grafting and were killed (b, c)

    5, 10, 15, or (d, e, f) 20 weeks post grafting. Ventral hair loss (a) was measured

    weekly, with 300 representing a full coat of hair. Data are shown as mean±SEM.

    Hematoxylin and eosin (H&E) slides were scored by JP Sundberg on a scale of

    0–4. Data are shown as percentage of each score for (b) epidermal hyperplasia,

    (c) anagen, (d) lymphocytes, (e) outer root sheath hyperplasia, and (f) follicular

    dystrophy. n¼ 8–11. *Significantly different from chow-fed mice, Po0.05,different letters are significantly different, Po0.05. ORS, outer root sheath.

    FJ Duncan et al.Retinoid Metabolism in Alopecia Areata

    338 Journal of Investigative Dermatology (2013), Volume 133

  • 10

    10

    0

    0

    10

    20

    30

    40

    0.0

    0.0

    0.5

    1.0

    1.5

    1.0

    2.0

    2.0

    3.0

    4.0

    5

    15

    200 IU4 IU12 IU28 IUChow

    0 IU4 IU12 IU28 IUChow

    0 IU4 IU12 IU28 IUChow

    A

    B

    AA A

    CB BC B B

    ABC AB ABB

    t10t15

    t15

    0 IU4 IU12 IU28 IUChow

    10

    0

    5

    15

    20

    10

    0

    5

    15

    20

    25

    No lesion

    AA lesion

    No lesionAA lesion

    No lesionAA lesion

    No lesionAA lesion

    ABABt5

    t5t5

    t5

    t5t5BC

    ABAB

    C

    A

    A

    C

    BBB

    C AA

    AAAA

    B

    t5

    t5

    t5

    t5

    t15t15

    t15

    t10

    t10

    t10

    t5

    t5

    A

    Ct10

    t10t15t15A

    ABAB

    BB

    B

    B

    B

    A

    A

    A A AA

    8

    6

    Num

    ber

    of G

    ZM

    B-p

    ositi

    vece

    llsN

    umbe

    r of

    GZ

    MB

    -pos

    itive

    cells

    Num

    ber

    of G

    ZM

    B-p

    ositi

    vece

    lls

    Num

    ber

    of G

    ZM

    B-p

    ositi

    vece

    lls

    Num

    ber

    of G

    ZM

    B-p

    ositi

    vece

    lls

    Num

    ber

    of G

    ZM

    B-p

    ositi

    vece

    lls

    Num

    ber

    of G

    ZM

    B-p

    ositi

    vece

    llsN

    umbe

    r of

    GZ

    MB

    -pos

    itive

    cells

    4

    2

    0

    10

    8

    6

    4

    2

    0

    5 10 15 20 5 10 15 20Week

    5 10 15 20

    Week

    5 10 15 20

    Week

    **

    *

    *

    *

    5 10 15 20Week

    Week

    5 10 15 20

    Week

    A

    A

    AB

    t10t10

    t10

    t15

    t10BB B B B

    BB

    B

    B B

    B

    5 10 15 20

    Week

    5 10 15 20Week

    **

    *

    *

    *

    *

    Figure 4. Granzyme B (GZMB) is significantly increased in mice with alopecia areata (AA) and altered by vitamin A. Immunohistochemistry (IHC) was

    performed and GZMB–positive cells enumerated in (a, b) the infundibulum, (c, d) isthmus, (e, f) suprabulbar, and (g, h) bulb. (a, c, e, g) Mann–Whitney tests were

    conducted to compare mice with AA lesions with mice without lesions (n¼40–50). (b, d, f, h) Generalized linear model with the Poisson response function wasthen performed on mice that received the AA graft with hair follicles in mid-anagen through mid-catagen (n¼ 1–10). Data are shown as mean±SE. *Po0.05versus no lesion; different letters within a time point are significantly different, Po0.05; t5, t10, t15 significantly different from 5, 10, and 15 weeks, respectively,Po0.05.

    4,000

    3,000

    200

    ba

    150

    100

    50

    05 10 15

    Week post grafting20

    2,000

    1,000

    IL13

    (pg

    per

    mg

    prot

    ein)

    IFN

    γ(p

    g pe

    r m

    g to

    tal p

    rote

    in)

    0Sham & AA

    AB

    Chow28 IU12 IU4 IU0 IU

    Chow28 IU12 IU4 IU0 IU

    ABAB

    ABAB

    AB

    ABAC ACAB

    AB ABABAB

    B

    B

    A

    C

    ABAB

    ABAB

    ABABB

    A

    Figure 5. Alterations in protein levels of cytokines by high vitamin A during alopecia areata (AA) progression in C3H/HeJ mice. (a) Protein levels of cytokines

    IL13 and (b) IFNg were determined by ELISA from dorsal lumbar skin. Data were natural log-(ln) transformed and analysis of variance performed using SPSS, v19.Data are shown as mean±SE. n¼ 63–65 for a, and n¼ 16–19 for b. Different letters are significantly different Po0.05.

    FJ Duncan et al.Retinoid Metabolism in Alopecia Areata

    www.jidonline.org 339

    http://www.jidonline.org

  • anagen transition (Shih et al., 2009). This increased anageninduction and alopecia could be reduced in Dgat1tm2Far-nullmice by severely reducing dietary vitamin A intake. In thisstudy, a moderate 3-fold increase in dietary vitamin Aaccelerated AA onset, possibly by inducing anagen, thetarget of AA. This dietary vitamin A level is well within theamount consumed by Americans who take supplements (Parket al., 2008). Thus, one mechanism for the increased AA withhigh vitamin A may be to regulate the hair cycle.

    Vitamin A may also directly regulate the immune response,as RA regulates numerous immune cells (Duriancik et al.,2010). Vitamin A increases Th2 and reduces Th1 cytokines(Hoag et al., 2002; Iwata et al., 2003). The current study foundthat high vitamin A reduced protein levels of the Th1 cytokineIFNg and increased Th2 cytokine IL13 over the control diet. Itis noteworthy that there are many differences between thesediets besides the vitamin A content. The reduction in IFNgmay be important, as it was shown to have a key role in theetiology of AA (Freyschmidt-Paul et al., 2005; Gilhar et al.,2005; Freyschmidt-Paul et al., 2006; Nakamura et al., 2008).Several studies reported increased skin IFNg mRNA levels,similar to the current finding (Carroll et al., 2002; McPheeet al., 2012). IFNg is regulated posttranscriptionally (Khabarand Young, 2007), yet few studies have measured IFNgprotein levels in AA. Serum IFNg protein levels wereincreased in patients with AA (Barahmani et al., 2009; Arcaet al., 2004). Within the skin of patients with long-standingAA, IFNg levels were reduced (Deeths et al., 2006). This isconsistent with the drop in skin IFNg found in the currentstudy. Collectively, the results from these studies suggest thatalthough IFNg is essential for AA onset skin IFNg levels maydrop as AA progresses. High vitamin A may accelerate AAtoward a chronic stage and associated reduced IFNg. Futurestudies should analyze IFNg protein levels in the serum,spleen, and/or lymph nodes.

    Vitamin A has a large role in maintaining gut immuneprivilege (Duriancik et al., 2010). RA synthesized in DCsincreased FOXP3þ Tregs and reduced Th17 cells to maintainmucosal immune tolerance. Increased Th17 cytokine levelsare hallmarks of autoimmune disease (Pelidou et al., 2000;Nakae et al., 2003; Langrish et al., 2005). The current reportshows that ALDH1A2 localized to dermal DCs, but Th17 cellsprobably do not contribute to AA in C3H/HeJ mice as cytokinelevels were reduced, not elevated. Th17 cytokines levels mayhave been too reduced for vitamin A to have any furthereffect. IL10, produced by numerous cell types (reviewed inO’Garra et al., 2008), is a key immunosuppressive cytokine inTreg differentiation (Asseman and Powrie, 1998; Assemanet al., 1999). IL10 can also activate CD8þ T cells (Grouxet al., 1998), and IL10 therapy produced variable clinicaloutcomes (O’Garra et al., 2008). AA frequency is reduced inIL10-deficient mice (Freyschmidt-Paul et al., 2002), suggestingthat IL10 may activate CD8þ T cells and exacerbate thedisease. Vitamin A may regulate IL10, but results areconflicting (Cantorna et al., 1994; Stephensen et al., 2004;Maynard et al., 2009). In the current report, IL10 was highestat 10 weeks, and then became significantly reduced as AAprogressed, but vitamin A had no effects. As IL10 has many

    roles, it may be immunosuppressive in sham mice butproinflammatory in AA mice. The reduction in IL10 at 20weeks in AA may point to a loss in function or a reduction inthe number of Tregs in AA. Other studies (Zoller et al., 2002;McElwee et al., 2005; Petukhova et al., 2010) provide indirectevidence that Tregs are important in AA, as IL2 and itsreceptor IL2A (CD25) were reduced in AA. IL2 is essentialfor the maintenance and survival of Treg cells (Fontenot et al.,2005). Overall, the results of this study suggest that althoughTh17 cells may not be involved in AA Tregs possibly are.There was no effect of dietary vitamin A on the immunefactors tested, but RA synthesis enzymes are localized to DCs.Future studies on AA should examine factors such as FOXP3and Treg function to further analyze the role(s) of vitamin A inTreg responses in AA.

    Both CTLA4 and PD1 (CD279) and its ligand PDL1 (CD274)are essential for suppressing T-cell responses directly andindirectly via Treg activation and preventing autoimmunediseases (Fife and Bluestone, 2008). PDL1 is expressed inmany immune privileged sites. To our knowledge, a reductionin PDL1 in AA that affected mouse hair follicles is previouslyunreported. PDL1 reduction later in disease is consistent withcompromised immune tolerance (privilege) in AA. In contrast,although CTLA4 decreased with time post grafting in AAaffected mouse skin, there were no significant differencesbetween AA and sham mice. These results conflict with geneassociation studies in humans (Petukhova et al., 2010; Johnet al., 2011) and functional studies in C3H/HeJ mice (Carrollet al., 2002; McElwee et al., 2002; Sundberg et al., 2011)linking CTLA4 to the pathogenesis of AA. One potentialreason for this difference may be the site tested. Skin wasexamined in this study, but CTLA4 alterations may occur inthe spleen and/or lymph nodes and should be examined infuture studies in these sites. The PD1 pathway, a secondcostimulatory pathway, should be examined for its role(s) inthe pathogenesis of AA.

    Increased GZMB expression occurs in numerous autoim-mune and skin diseases (Boivin et al., 2009; Afonina et al.,2010). GZMB was localized to the dermis, subcutis, and hairfollicle connective tissue sheath in some but not all studies onAA patients (Sato-Kawamura et al., 2003). Small numbers ofGZMB-positive cells were detected in the dermis of C3H/HeJmice and were increased by tachykinin 1 (substance P,(Siebenhaar et al., 2007). None of these studies reported theGZMB-positive cell localization within the hair follicle. In thecurrent study, GZMB-positive cells were detected around andwithin the hair follicle, as well as in the isthmus—the site ofhair follicle stem cells (Cotsarelis et al., 1990) and immuneprivilege collapse in AA (Meyer et al., 2008). Most studiesonly mention GZMB’s role in inducing apoptosis, and someargue against GZMB having a major role in killing follicularcells in AA. More recent studies reveal additional roles forGZMB in cleaving extracellular matrix proteins, autoantigens,and receptors NOTCH1 and FGFR1 (Boivin et al., 2009).These effects could negatively affect the hair follicle in AA byaltering both the stucture of the connective tissue sheath andsignaling within the hair follicle stem cells and dermal papilla.Breakdown of the extracellular matrix can cause cell death

    FJ Duncan et al.Retinoid Metabolism in Alopecia Areata

    340 Journal of Investigative Dermatology (2013), Volume 133

  • because of loss of cell–matrix interactions, allow moreimmune cells access to follicles, and contribute to collapseof hair follicle immune privilege. Thus, altered immunoloca-lization of GZMB by vitamin A may cause different types ofcellular damage at different follicular sites.

    The chemokines tested in this study increased consistentlywith recruitment of immune cells (Hayashi, 1982; Shimokawaet al., 1983; Rathanaswami et al., 1993; Flier et al., 2001). Theincrease in CCL5 and CXCL9 in C3H/HeJ mice is similar toreported increases in gene and protein expression (Kuwanoet al., 2007; Subramanya et al., 2010) in patients with AA. Thecolocalization of RA synthesis enzymes with NKG2D cellssuggests that AA may be altered by RA effects on these cells.RA upregulates several NKG2D ligands (Cerwenka et al.,2000; Jinushi et al., 2003; Poggi et al., 2009), which activateNK, NKT, and CD8þ T cells (Mistry and O’Callaghan, 2007),but whether and how vitamin A regulates NKG2D and itsligands is unknown.

    Initial attempts to treat AA with retinoids gave promisingresults. Topical tretinoin (0.05%) alone (Much, 1976), orcombined with intralesional triamcinolone acetonide(Kubeyinje and Cmathur, 1997), was effective in somepatients. The RXR agonist, bexarotene, also induced hairregrowth in some patients (Talpur et al., 2009). None ofthese studies had more than 42 patients, nor were theycontrolled for spontaneous hair regrowth. In the currentstudy, high vitamin A accelerated the disease, arguingagainst pharmacologic retinoids as an effective treatment forAA. It is also possible that the patients who responded toretinoid treatment were marginally vitamin A deficient andthat treatment corrected this deficiency. AA waxes and wanes;therefore, the stage of disease at the time of treatment mayalso alter retinoid effectiveness. In addition, studies in the gutsuggest that exogenous retinoids do not alter Tregs (Xiao et al.,2008), whereas upregulating RA within DCs did (Housleyet al., 2009; Manicassamy et al., 2009), suggesting thatregulating DC RA production may be an effective treatment.This study also highlights the need for precise regulation of RAin AA patients with adequate dietary vitamin A, as excessvitamin A from physicians and commercial sources such asskin- and hair-care products (Arechalde and Saurat, 2000) anddietary supplements (Park et al., 2008) may be detrimental.

    MATERIALS AND METHODSAll procedures were carried out with approval from The Jackson

    Laboratory IACUC for the microarray study, The Ohio State Uni-

    versity IACUC for the diet study, and both IACUC boards for the mice

    used for retinoid analysis. The microarray study was previously

    reported (McPhee et al., 2012). Tissue arrays were created from

    archived tissues (Sundberg et al., 1994, 1995; McElwee et al., 1998b,

    1999). Biopsies from human patients (n¼ 2) were obtained fromarchives at Vanderbilt University (Nashville, TN) and Baylor Hair

    Research and Treatment Center (Dallas, TX, Supplementary Table S1

    online). The Vanderbilt University Institutional Review Board

    approved all human work. Rat paraffin blocks of skin with or without

    AA (n¼ 3) were provided by Kevin McElwee at the University ofBritish Columbia. IHC and immunoflurescence were performed as

    previously described (Everts et al., 2007; Duncan et al., 2009).

    Primary antibodies against Langerin (eBiosciences, San Diego, CA),

    NKG2D, GZMB, and PDL1 (CD274; R&D, Minneapolis, MN) were

    purchased, whereas the remaning antibodies were prepared and

    validated in Dr Ong’s laboratory (Everts et al., 2007) (Supplementary

    Table S2 online).

    In the second study, recipient mice (n¼ 10) were fed purifiedAIN93M diets containing either 0 (VAD), 4 (VAA), 12 (VAH), or 28

    (VAE) IU retinyl acetate per g diet (Research Diets, New Brunswick,

    NJ), or control unpurified diet (chow, Harlen Teklad 7912, Indiana-

    polis, IN), starting 2 weeks before grafting according to McElwee et al.

    (1998a). Body weight, food intake, and hair loss were measured (Tang

    et al., 2004) weekly and samples collected 5, 10, 15, and 20 weeks

    post grafting. Histology was scored by JPS (Supplementary Figure S1

    online). ELISA kits were purchased from R&D Systems, whereas the

    ELISA array was purchased from SABiosciences (Frederick, MD), and

    analyses were performed according to the kit instructions. RNA was

    isolated using the RNAeasy Fibrous tissue kit (Qiagen, Valencia, CA),

    cDNA prepared using the High Capacity cDNA Archive Kit (Applied

    Biosystems, Carlsbad, CA), and quantitative real-time PCR performed

    using a Taqman kit (Applied Biosystems).

    Additional AA and sham mice (n¼ 6) were obtained (JPS) andretinoids analyzed as previously described (Kane et al., 2008a, b).

    Data were analyzed using SPSS, v19 (IBM; Armonk, NY) after

    consultation with the OSU statistical consulting services. Histological

    scores were analyzed using a generalized linear model with the

    multinomial logistic response function at each time point. See the

    figure legends for additional analyses.

    See Supplementary Data online for more details.

    CONFLICT OF INTERESTThe authors state no conflict of interest.

    ACKNOWLEDGMENTSThis work was supported by grants from the National Alopecia AreataFoundation (to HBE, LEK, and JPS) and the National Institutes of Health(AR052009 to HBE, AR041943 to LEK, and AR056635 to JPS). We thank JeniSquiric and Steven Naber from the OSU statistical department for their guidance.

    SUPPLEMENTARY MATERIAL

    Supplementary material is linked to the online version of the paper at http://www.nature.com/jid

    REFERENCES

    Afonina IS, Cullen SP, Martin SJ (2010) Cytotoxic and non-cytotoxic roles of

    the CTL/NK protease granzyme B. Immunol Rev 235:105–16

    Anzano MA, Lamb AJ, Olson JA (1979) Growth, appetite, sequence of

    pathological signs and survival following the induction of rapid, synchro-

    nous vitamin A deficiency in the rat. J Nutr 109:1419–31

    Arca E, Musabak U, Akar A et al. (2004) Interferon-gamma in alopecia areata.

    Eur J Dermatol 14:33–6

    Arechalde A, Saurat JH (2000) Retinoids: unapproved uses or indications. Clin

    Dermatol 18:63–76

    Asseman C, Mauze S, Leach MW et al. (1999) An essential role for interleukin

    10 in the function of regulatory T cells that inhibit intestinal inflammation.

    J Exp Med 190:995–1004

    Asseman C, Powrie F (1998) Interleukin 10 is a growth factor for a population

    of regulatory T cells. Gut 42:157–8

    Balmer JE, Blomhoff R (2002) Gene expression regulation by retinoic acid.

    J Lipid Res 43:1773–808

    FJ Duncan et al.Retinoid Metabolism in Alopecia Areata

    www.jidonline.org 341

    http://www.nature.com/jidhttp://www.nature.com/jidhttp://www.jidonline.org

  • Barahmani N, Lopez A, Babu D et al. (2009) Serum T helper 1 cytokine levelsare greater in patients with alopecia areata regardless of severity of atopy.Clin Exp Dermatol 35:409–16

    Boivin WA, Cooper DM, Hiebert PR et al. (2009) Intracellular versusextracellular granzyme B in immunity and disease: challenging thedogma. Lab Invest 89:1195–220

    Bursch LS, Wang L, Igyarto B et al. (2007) Identification of a novel populationof Langerinþ dendritic cells. J Exp Med 204:3147–56

    Cantorna MT, Nashold FE, Hayes CE (1994) In vitamin A deficiency multiplemechanisms establish a regulatory T-helper cell imbalance with excessTh1 and insufficent Th2 function. J Immunol 152:1515–22

    Carroll JM, McElwee KJ, King LEJ et al. (2002) Gene array profiling andimmunomodulation studies define a cell-mediated immune responseunderlying the pathogenesis of alopecia areata in a mouse model andhumans. J Invest Dermatol 119:392–402

    Cerwenka A, Bakker ABH, McClanahan T et al. (2000) Retinoic acid earlyinducible genes define a ligand family for the activating NKG2D receptorin mice. Immunity 12:721–7

    Coombes JL, Siddiqui KRR, Arancibia-Carcamo CV et al. (2007) A functionallyspecialized population of mucosal CD103þ DCs induces Foxp3þ

    regulatory T cells via a TGF-beta- and retinoic acid-dependent mechan-ism. J Exp Med 204:1757–64

    Cotsarelis G, Sun TT, Lavker RM (1990) Label-retaining cells reside in the bulgearea of pilosebaceous unit: implications for follicular stem cells, haircycle, and skin carcinogenesis. Cell 61:1329–37

    Deeths MJ, Endrizzi BT, Irvin ML et al. (2006) Phenotypic analysis of T-cells inextensive alopecia areata scalp suggests partial tolerance. J InvestDermatol 126:366–73

    Duncan FJ, Martin JR, Wulff BC et al. (2009) Topical treatment with blackraspberry extract reduces cutaneous uvb-induced carcinogenesis andinflammation. Cancer Prev Res 2:665–72

    Duriancik DM, Lackey DE, Hoag KA (2010) Vitamin A as a regulator of antigenpresenting cells. J Nutr 140:1395–9

    Everts HB (2012) Endogenous retinoids in the hair follicle and sebaceous gland.Biochim Biophys Acta 1821:222–9

    Everts HB, Berdanier CD (2002) Nutrient-gene interactions in mitochondrialfunction: vitamin A needs are increased in BHE/Cdb rats. IUBMB Life53:289–94

    Everts HB, Sundberg JP, King LE Jr. et al. (2007) Immunolocalization ofenzymes, binding proteins, and receptors sufficient for retinoic acidsynthesis and signaling during the hair cycle. J Invest Dermatol127:1593–604

    Fife BT, Bluestone JA (2008) Control of peripheral T-cell tolerance andautoimmunity via the CTLA-4 and PD-1 pathways. Immunol Rev224:166–82

    Flier J, Boorsma DM, van Beek PJ et al. (2001) Differential expression ofCXCR3 targeting chemokines CXCL10, CXCL9, and CXCL11 in differenttypes of skin inflammation. J Pathol 194:398–405

    Flowers MT, Paton CM, O’Byrne SM et al. (2011) Metabolic changes in skincaused by Scd1 deficiency: a focus on retinol metabolism. PLoS One6:e19734

    Fontenot JD, Rasmussen JP, Gavin MA et al. (2005) A function for interleukin 2in Foxp3-expressing regulatory T cells. Nat Immunol 6:1142–51

    Freyschmidt-Paul P, McElwee KJ, Happle R et al. (2002) Interleukin-10-deficient mice are less susceptible to the induction of alopecia areata.J Invest Dermatol 119:980–2

    Freyschmidt-Paul P, McElwee KJ, Hoffmann R et al. (2005) Reduced expressionof interleukin-2 decreases the frequency of alopecia areata onset in C3H/HeJ mice. J Invest Dermatol 125:945–51

    Freyschmidt-Paul P, McElwee KJ, Hoffmann R et al. (2006) Interferon-gamma-deficient mice are resistant to the development of alopecia areata. Br JDermatol 155:515–21

    Gilhar A, Kam Y, Assy B et al. (2005) Alopecia areata induced in C3H/HeJmice by interferon-gamma: evidence for loss of immune privilege. J InvestDermatol 124:288–9

    Girard C, Dereure O, Blatiere V et al. (2006) Vitamin A deficiencyphrynoderma associated with chronic giardiasis. Pediatr Dermatol23:346–9

    Groux H, Bigler M, de Vries JE et al. (1998) Inhibitory and stimulatory effects ofIL-10 on human CD8þ T cells. J Immunol 160:3188–93

    Guilliams M, Crozat K, Henri S et al. (2010) Skin-draining lymph nodescontain dermis-derived CD103� dendritic cells that constitutively pro-duce retinoic acid and induce Foxp3þ regulatory T cells. Blood115:1958–68

    Harries MJ, Sun J, Paus R et al. (2010) Management of alopecia areata. BMJ341:c3671

    Hayashi H (1982) A review on the natural mediators of inflammatoryleucotaxis. Acta Pathol Jpn 32(Suppl 2):271–84

    Hoag KA, Nashold FE, Goverman J et al. (2002) Retinoic acid enhancesthe T helper 2 cell development that is essential for robust anti-body responses through its action on antigen-presenting cells. J Nutr132:3736–9

    Housley WJ, O’Conor CA, Nichols F et al. (2009) PPAR gamma regulatesretinoic acid-mediated DC induction of Tregs. J Leukoc Biol 86:293–301

    Ito T, Ito N, Saatoff M et al. (2008) Maintenance of hair follicle immuneprivilege is linked to prevention of NK cell attack. J Invest Dermatol128:1196–206

    Iwata M, Eshima Y, Kagechika H (2003) Retinoic acids exert direct effects on Tcells to suppress Th1 development and enhance Th2 development viaretinoic acid receptors. Int Immunol 15:1017–25

    Iwata M, Hirakiyama A, Eshima Y et al. (2004) Retinoic acid imprints gut-homing specificity on T cells. Immunity 21:527–38

    Jinushi M, Takehara T, Tatsumi T et al. (2003) Expression and role of MICA andMICB in human hepatocellular carcinomas and their regulation byretinoic acid. Int J Cancer 104:354–61

    John KK, Brockschmidt FF, Redler S et al. (2011) Genetic variants inCTLA4 are strongly associated with alopecia areata. J Invest Dermatol131:1169–72

    Kane MA, Folias AE, Napoli JL (2008a) HPLC/UV quantitation of retinal,retinol, and retinyl esters in serum and tissues. Anal Biochem 378:71–9

    Kane MA, Folias AE, Wang C et al. (2008b) Quantitative profiling ofendogenous retinoic acid in vivo and in vitro by tandem mass spectro-metry. Anal Chem 80:1702–8

    Khabar KSA, Young HA (2007) Post-transcriptional control of the interferonsystem. Biochimie 89:761–9

    King LE, McElwee KJ, Sundberg JP (2008) Alopecia Areata. In: Nickoloff BJ,Nestle FO (eds) Dermatologic Immunity, Curr Dir Autoimmun. Basel:Karger, 280–312

    Kubeyinje EP, Cmathur M (1997) Topical tretinoin as an adjunctive therapywith intralesional triamcinolone acetonide for alopecia areata. Clinicalexperience in northern Saudi Arabia. Int J Dermatol 36:320

    Kuwano Y, Fujimoto M, Watanabe R et al. (2007) Serum chemokine profiles inpatients with alopecia areata. Br J Dermatol 157:466–73

    Langrish CL, Chen Y, Blumenschein WM et al. (2005) IL-23 drives apathogenic T cell population that induces autoimmune inflammation.J Exp Med 201:233–40

    Manicassamy S, Ravindran R, Deng JS et al. (2009) Toll-like receptor2-dependent induction of vitamin A-metabolizing enzymes in dendriticcells promotes T regulatory responses and inhibits autoimmunity. NatMed 15:401–9

    Maynard CL, Hatton RD, Helms WS et al. (2009) Contrasting roles for all-transretinoic acid in TGF-beta-mediated induction of Foxp3 and Il10 genes indeveloping regulatory T cells. J Exp Med 206:343–57

    McElwee KJ, Boggess D, King LE Jr et al. (1998a) Experimental induction ofalopecia areata-like hair loss in C3H/HeJ mice using full-thickness skingrafts. J Invest Dermatol 111:797–803

    McElwee KJ, Boggess D, Miller J et al. (1999) Spontaneous alopecia areata-likehair loss in one congenic and seven inbred laboratory mouse strains.J Invest Dermatol Symp Proc 4:202–6

    McElwee KJ, Boggess D, Olivry T et al. (1998b) Comparison of alopecia areatain human and nonhuman mammalian species. Pathobiology 66:90–107

    FJ Duncan et al.Retinoid Metabolism in Alopecia Areata

    342 Journal of Investigative Dermatology (2013), Volume 133

  • McElwee KJ, Freyschmidt-Paul P, Hoffmann R et al. (2005) Transfer ofCD8þ cells induces localized hair loss whereas CD4þ /CD25�

    cells promote systemic alopecia areata and CD4þ /CD25þ cellsblockade disease onset in the C3H/HeJ mouse model. J Invest Dermatol124:947–57

    McElwee KJ, Hoffmann R, Freyschmidt-Paul P et al. (2002) Resistance toalopecia areata in C3H/HeJ mice is associated with increased expressionof regulatory cytokines and a failure to recruit CD4þ and CD8þ cells.J Invest Dermatol 119:1426–33

    McElwee KJ, Spiers EM, Oliver RF (1996) In vivo depletion of CD8þ T cellsrestores hair growth in the DEBR model for alopecia areata. Br J Dermatol135:211–7

    McPhee CG, Duncan FJ, Silva KA et al. (2012) Increased expression of Cxcr3and its ligands, Cxcl9 and Cxcl10, during the development of alopeciaareata in the mouse. J Invest Dermatol 132:1736–8

    Meyer KC, Klatte JE, Dinh HV et al. (2008) Evidence that the bulge region is asite of relative immune privilege in human hair follicles. Br J Dermatol159:1077–85

    Mistry AR, O’Callaghan CA (2007) Regulation of ligands for the activatingreceptor NKG2D. Immunology 121:439–47

    Much T (1976) Treatment of alopecia areata with vitamin A acid. Z Hautkr51:993–8

    Mucida D, Park Y, Kim G et al. (2007) Reciprocal TH17 and regulatory T celldifferentiation mediated by retinoic acid. Science 317:256–60

    Nakae S, Saijo S, Horai R et al. (2003) IL-17 production from activated T cellsis required for the spontaneous development of destructive arthritis inmice deficient in IL-1 receptor antagonist. Proc Natl Acad Sci USA100:5986–90

    Nakamura M, Jo J, Tabata Y et al. (2008) Controlled delivery of T-box21 smallinterfering RNA ameliorates autoimmune alopecia (Alopecia Areata) in aC3H/HeJ mouse model. Am J Pathol 172:650–8

    O’Garra A, Barrat FJ, Castro AG et al. (2008) Strategies for use of IL-10 or itsantagonists in human disease. Immunol Rev 223:114–31

    Park SY, Murphy SP, Martin CL et al. (2008) Nutrient intake from multivitamin/mineral supplements is similar among users from five ethnic groups: TheMultiethnic Cohort Study. J Am Diet Assoc 108:529–33

    Pelidou SH, Zou LP, Deretzi G et al. (2000) Enhancement of acute phase andinhibition of chronic phase of experimental autoimmune neuritis in Lewisrats by intranasal administration of recombinant mouse interleukin 17:potential immunoregulatory role. Exp Neurol 163:165–72

    Petukhova L, Duvic M, Hordinsky M et al. (2010) Genome-wide associationstudy in alopecia areata implicates both innate and adaptive immunity.Nature 466:113–7

    Poggi A, Catellani S, Garuti A et al. (2009) Effective in vivo induction ofNKG2D ligands in acute myeloid leukaemias by all-trans-retinoic acid orsodium valproate. Leukemia 23:641–8

    Rathanaswami P, Hachicha M, Sadick M et al. (1993) Expression of thecytokine RANTES in human rheumatoid synovial fibroblasts. Differentialregulation of RANTES and interleukin-8 genes by inflammatory cytokines.J Biol Chem 268:5834–9

    Ries G, Hess R (1999) Retinol: Safety considerations for its use in cosmeticproducts. J Toxicol Cutaneous Ocul Toxicol 18:169–85

    Ruzicka T, Sommerburg C, Goerz G et al. (1992) Treatment of cutaneouslupus-erythematosus with acitretin and hydroxychloroquine. Br J Derma-tol 127:513–8

    Safavi KH, Muller SA, Suman VJ et al. (1995) Incidence of alopecia areata inOlmsted County, Minnesota, 1975 through 1989. Mayo Clin Proc70:628–33

    Sato-Kawamura M, Aiba S, Tagami H (2003) Strong expression of CD40, CD54and HLA-DR antigen and lack of evidence for direct cellular cytotoxicityare unique immunohistopathological features in alopecia areata. ArchDermatol Res 294:536–43

    Schambach F, Schupp M, Lazar MA et al. (2007) Activation of retinoic acidreceptor-alpha favours regulatory T cell induction at the expense of IL-17-secreting T helper cell differentiation. Eur J Immunol 37:2396–9

    Shih MYS, Kane MA, Zhou P et al. (2009) Retinol esterification by DGAT1is essential for retinoid homeostasis in murine skin. J Biol Chem284:4292–9

    Shimokawa Y, Miura K, Hifumi M et al. (1983) Lymphocyte chemotaxis ininflammation. VI. Lyt phenotype analysis of effector cells responsible forproducing murine lymphocyte chemotactic factor. Immunology 49:95–102

    Siebenhaar F, Sharov AA, Peters EM et al. (2007) Substance P as animmunomodulatory neuropeptide in a mouse model for autoimmunehair loss (alopecia areata). J Invest Dermatol 127:1489–97

    Smith SM, Levy NS, Hayes CE (1987) Impaired immunity in vitamin A deficientmice. J Nutr 117:857–65

    Sojka DK, Huang YH, Fowell DJ (2008) Mechanisms of regulatoryT-cell suppression—a diverse arsenal for a moving target. Immunology124:13–22

    Stephensen CB, Jiang XW, Freytag T (2004) Vitamin A deficiency increases thein vivo development of IL-10-positive Th2 cells and decreases develop-ment of Th1 cells in mice. J Nutr 134:2660–6

    Stockinger B, Veldhoen M, Martin B (2007) Th17 T cells: linking innate andadaptive immunity. Sem Immunol 19:353–61

    Subramanya RD, Coda AB, Sinha AA (2010) Transcriptional profiling inalopecia areata defines immune and cell cycle control related geneswithin disease-specific signatures. Genomics 96:146–53

    Sundberg JP, Cordy WR, King LE Jr (1994) Alopecia areata in aging C3H/HeJmice. J Invest Dermatol 102:847–56

    Sundberg JP, McElwee KJ, Carroll JM et al. (2011) Hypothesis testing: CTLA4co-stimulatory pathways critical in the pathogenesis of human and mousealopecia areata. J Invest Dermatol 131:2323–4

    Sundberg JP, Oliver RF, McElwee KJ et al. (1995) Alopecia areata in humansand other mammalian species. J Invest Dermatol 104:32S–3S

    Sundberg JP, Silva KA, Li RH et al. (2004) Adult-onset alopecia areata is acomplex polygenic trait in the C3H/HeJ mouse model. J Invest Dermatol123:294–7

    Talpur R, Vu J, Bassett R et al. (2009) Phase I/II randomized bilateral half-headcomparison of topical bexarotene 1% gel for alopecia areata. J Am AcadDermatol 61:592–8

    Tang L, Cao L, Sundberg JP et al. (2004) Restoration of hair growth in mice withan alopecia areata-like disease using topical anthralin. Exp Dermatol13:5–10

    Tosti A, Duque-Estrada B (2009) Treatment strategies for alopecia. Expert OpinPharmacother 10:1017–26

    Wolbach SB, Howe PR (1925) Tissue changes following deprivation of fat-soluble A vitamin. J Exp Med 42:753–77

    Xiao S, Jin H, Korn T et al. (2008) Retinoic acid increases Foxp3þ regulatory Tcells and inhibits development of Th17 cells by enhancing TGF-beta-driven Smad3 signaling and inhibiting IL-6 and IL-23 receptor expression.J Immunol 181:2277–84

    Zoller M, McElwee KJ, Engel P et al. (2002) Transient CD44 variant isoformexpression and reduction in CD4þ /CD25þ regulatory T cells in C3H/HeJmice with alopecia areata. J Invest Dermatol 118:983–92

    FJ Duncan et al.Retinoid Metabolism in Alopecia Areata

    www.jidonline.org 343

    http://www.jidonline.org

    Endogenous Retinoids in the Pathogenesis of Alopecia AreataIntroductionResultsDiscussionMaterials and Methods