You are on page 1of 6

REVIEW

Hair Follicle Immune Privilege Revisited:


The Key to Alopecia Areata Management
Ralf Paus1, Silvia Bulfone-Paus1 and Marta Bertolini2
The collapse of the immune privilege (IP) of the ana- state established by complex tissue-specific mechanisms that
gen hair bulb is now accepted as a key element in AA suppress inflammation and promote immune tolerance
pathogenesis, and hair bulb IP restoration lies at the (Engelhardt et al., 2017). Although the few tissues that enjoy
core of AA therapy. Here, we briefly review the es- IP differ in their IP state and characteristics, the most crucial
sentials of hair bulb IP and recent progress in under- common mechanisms shared by them are (i) low or absent
standing its complexity. We discuss open questions expression of major histocompatibility complex (MHC) class
and why the systematic dissection of hair bulb IP and Ia/b2 microglobulin expression, thus rendering self-peptide
its pharmacological manipulation (including the clin- presentation ineffective or impossible, and (ii) the creation of
ical testing of FK506 and a-melanocyte-stimulating an immunoinhibitory milieu by the generation of secreted
immunosuppressants (Engelhardt et al., 2017; Joyce and
hormone analogs) promise to extend the range of
Fearon, 2015; Kinori et al., 2011; Nasr et al., 2005; Noso
future therapeutic options in AA and other IP collapse-
et al., 2015; Paus et al., 2005; Taylor, 2016). Also, IP tissues
related autoimmune diseases.
can induce a state of tolerance against antigens that manage to
Journal of Investigative Dermatology Symposium Proceedings (2018) 19, escape immune sequestration (Taylor, 2016) and down-
S12eS17; doi:10.1016/j.jisp.2017.10.014
regulate T-cell activation and proliferation (Eleftheriadis
et al., 2016; Ma et al., 2017; Tan and Bharath, 2009).
All of these mechanisms are established in the hair bulge IP
HAIR FOLLICLE IMMUNE PRIVILEGE: INCEPTION AND and hair bulb IP of murine and human HFs (Christoph et al.,
BASICS 2000; Harrist et al., 1983; Meyer et al., 2008; Paus et al.,
Rupert Billingham recognized in the skin of guinea pigs that 2005), with the bulb IP being the one that is relevant to AA
transplanted heterologous epidermal melanocytes can pathogenesis. Although the functions of HF IP remain spec-
escape immune elimination when they migrate to the anagen ulative, current evidence suggests that the main function of
hair bulb epithelium of genetically incompatible host hair the anagen hair bulb IP may be to sequester immunogenic,
follicles (HFs) (Billingham and Silvers, 1971). This striking melanogenesis-associated, and/or other HF antigens pro-
finding was largely ignored until Gill Westgate recalled duced in the anagen hair bulb from immune recognition
attention to the concept of HF immune privilege (IP) (Paus et al., 1993, 2005). With increasing insights into the
(Westgate et al., 1991). This encouraged the proposal that immunological features of the HF over the past decade, the
hair bulb IP collapse constitutes a crucial element in HF understanding of HF IP has attained new layers of complexity
pathobiology, namely in alopecia areata (AA) (Paus et al., (Bertolini et al., 2016; Breitkopf et al., 2013; Ito and Tokura,
1993) and triggered a series of studies to better define HF 2014; Kang et al., 2010; Kinori et al., 2012; Paus et al., 2005;
immunology (e.g., Christoph et al., 2000; Hoffman et al., Wang et al., 2014). These, and potential mechanisms for how
1996; Paus et al., 1994a, 1994b, 1994c, 1998, 2005; Wang hair bulb IP is actively maintained, is threatened, collapses,
et al., 2014). and may best be repaired during AA management are dis-
The central role of IP collapse in AA pathobiology has now cussed below.
become widely accepted in the field (Gilhar et al., 1998;
Gilhar et al., 2012, 2016; Kang et al., 2010; McElwee AA AS A STEREOTYPIC HAIR FOLLICLE RESPONSE
et al., 2013; Pratt et al., 2017; Wang et al., 2014), and PATTERN
formal functional proof that anagen HFs do exhibit a relative AA typically begins with focal hair loss in uninflamed skin,
IP has been provided in mouse models (Alli et al., 2012; Gao which in some individuals progresses to universal hair loss.
et al., 2017; Giangreco et al., 2012). Histologically, AA lesions show a dense inflammatory infil-
Since its first inception by Peter Medawar (1948) the term trate around the hair bulb of melanogenically active anagen
IP has been extended to reflect a dynamic immunoinhibitory HFs (Miteva et al., 2012). This autoaggressive infiltrate causes
both premature termination of anagen, forcing the HF into
1 catagen (Oh et al., 2016), and major HF dystrophy (Gilhar
Centre for Dermatology Research, University of Manchester, NIHR
Manchester Biomedical Research Centre and MAHSC, Manchester, UK; et al., 2012). Therefore, AA represents both an HF cycling
and 2Department of Dermatology, University of Münster, Münster, and a hair growth disorder.
Germany The preference of AA for fully pigmented anagen VI HFs
Correspondence: Ralf Paus, Centre for Dermatology Research, University of has been used to explain the sparing of white HFs and has
Manchester, NIHR Manchester Biomedical Research Centre and MAHSC,
Stopford Building, Oxford Road, Manchester, M13 9PR, UK. E-mail: ralf.
invited the hypothesis that melanogenesis-associated auto-
paus@manchester.ac.uk antigens are the chief targets in AA (Paus et al., 1993). In AA,
Abbreviations: AA, alopecia areata; HF, hair follicle; IP, immune privilege; these autoantigens are most likely to be presented by
MHC, major histocompatibility complex; NK, natural killer MHC class Iaþ cells in the anagen hair bulb epithelium

ª 2017 The Authors. Published by Elsevier, Inc. on behalf of the


S12 Journal of Investigative Dermatology Symposium Proceedings (2018), Volume 19 Society for Investigative Dermatology.
R Paus et al.
Immune Privilege in Alopecia Areata

(Bertolini et al., 2014, 2016, 2017; Christoph et al., 2000; Ito differences in susceptibility to AA, in course of the hair loss
et al., 2004; Kinori et al., 2012). phenotype, in response to AA therapy, and in likelihood of
It has long been recognized that AA reflects a wide spec- spontaneous remission (Gilhar et al., 2012) may very well
trum of hair loss phenotypes (Gilhar et al., 2012; Ikeda, 1965; reflect constitutive, genetically determined differences in
Tosti et al., 2006). Moreover, AA clinical subtypes show some how easily the hair bulb IP collapses and how effectively HFs
shared gene associations, fitting the model of AA as a poly- manage to repair their IP.
genic disease with different presentations arising from a
combination of genetic and environmental factors (Betz HF IP RESTORATION AS THE CORNERSTONE OF
et al., 2015; Fischer et al., 2016; Petukhova et al., 2010). SUCCESSFUL AA MANAGEMENT
However, a phenocopy of AA lesions can be experimentally Thus, if one manages to protect and restore HF IP more
induced in healthy human scalp skin xenotransplants onto effectively, AA progression can be stopped, and spontaneous
SCID mice under conditions where an antigen-specific hair regrowth is predicted to occur, because the HF damage
autoimmune attack on the anagen HF and a genetic predis- associated with AA remains fully reversible.
position to AA are quite unlikely (Gilhar et al., 2013, 2016). However, the current standard therapy for AA (Kassira
Therefore, we have argued that AA should be considered et al., 2017) may do little to restore HF IP. Although un-
as a stereotypic response pattern that any anagen HFs will equivocal placebo-controlled evidence that JAK/STAT in-
show, irrespective of the genetic predisposition and the pre- hibitors are effective in AA therapy is not yet available, one
existence of autoreactive CD8þ T cells, provided that HF IP main reason why these therapeutics promote hair regrowth in
collapses and that excessive IFN-g signaling causes cytotoxic many, though not all, AA patients (Alves de Medeiros et al.,
HF damage (Paus, in: McElwee et al., 2013). According to 2016; Jabbari et al., 2015; Kennedy Crispin et al., 2016;
this concept, only a certain percentage of AA patients reflects Liu et al., 2017; Mackay-Wiggan et al., 2016) may be
the presence of an (auto)antigen-specific and CD8/NKG2D- because they block IFN-gemediated signaling (Xing et al.,
dependent autoimmune disease, which is best termed auto- 2014) and thus the promotion of HF IP collapse, besides
immune AA (AAA). Instead, other AA patients—perhaps affecting HF cycling (Harel et al., 2015).
those with the greatest likelihood of spontaneous remission— The relapse of hair loss often seen after AA patients have
may mainly exhibit the hair loss-triggering consequences of discontinued therapy with these promising new therapeutics
nonspecific, IFN-geinduced HF IP collapse, dystrophy, and and the development of refractoriness to this therapy in some
premature catagen (Ito et al., 2005); therefore, AA patients patients over time (Kennedy Crispin et al., 2016; Mackay-
may require personalized medicine management strategies Wiggan et al., 2016) underscore the ultimate goal of any
(Paus, in: McElwee et al., 2013). successful AA therapy, that is, to help the HF restore its IP—
the most effective safeguarding mechanism against AA
HF IP COLLAPSE AS THE CENTRAL PREREQUISITE OF THE relapse. Moreover, long-term therapy with potent JAK/STAT
AA HAIR LOSS PHENOTYPE inhibitors, which block T- and natural killer (NK)-cell func-
Regardless of whether or not AA is only a stereotypic HF tions (Xing et al., 2014), must be expected to severely
response pattern, AA cannot develop without the following compromise intracutaneous tumor immunosurveillance and
(Gilhar et al., 2012): the defense against viral infection—concerns that are not
circumvented by topical drug application (Kostovic et al.,
 the occurrence of a perifollicular inflammation around the 2017). Therefore, caution is still advised regarding the long-
anagen hair bulb; term administration of JAK/STAT inhibitors to AA patients.
 the induction of HF dystrophy, which leads to hair shaft From a drug safety and IP restoration efficiency perspec-
breakage or shedding and/or production of a dysfunctional tive, well-documented “IP guardians” such as synthetic
hair shaft; and, most important, a-MSH analogs (melanotan, alfamelanotide) need to be tested
 hair bulb IP collapse. clinically in AA. These not only potently down-regulate MHC
class Ia/b2 microglobulin expression in human anagen HFs
The bulb IP exists only in anagen, because most of the (Ito et al., 2004), but they may also stimulate production of
MHC class Ia/b2 microglobulin-negative HF keratinocytes other HF IP guardians, besides exerting direct immunoinhibi-
are deleted via apoptosis during catagen (Paus et al., 1994a, tory effects themselves (Brzoska et al., 2008; Chang et al.,
2005). Therefore, once an inflammation-damaged anagen HF 2008). Although this strategy has long been advocated
(Ito et al., 2004, 2005; Peters et al., 2007; Zarbo et al., 2017) (Gilhar et al., 2012; Ito et al., 2004) and despite the intro-
has entered into catagen and has resided in telogen (Oh duction of a-MSH analogs into clinical dermatology, industry
et al., 2016), it has the chance to completely reconstruct its has not yet advanced these agents for clinical testing in AA.
bulb IP with entry into the next anagen phase—unless the Although glucocorticoids suppress T-cell functions, MHC
newly developing anagen HF is attacked again. class II expression, and many proinflammatory signaling
Circumstantial evidence suggests that spontaneous resto- pathways, they are much less effective in down-regulating
ration of HF IP is promoted by the intrafollicular production MHC class Ia/b2 microglobulin expression (Truckenmiller
of endogenous IP guardians by outer root sheath keratino- et al., 2005) and stimulating the production of HF IP guard-
cytes (Breitkopf et al., 2013; Ito et al., 2004; Paus et al., 2005) ians, possibly explaining why glucocorticoids often disap-
and the release of immunoinhibitory neuropeptides from point in AA management.
perifollicular sensory nerve fibers (Bertolini et al., 2016; Instead, FK506 (tacrolimus), ideally in a topical prepara-
Kinori et al., 2012). Therefore, the interindividual tion that facilitates penetration and drug accumulation

www.jidsponline.org S13
R Paus et al.
Immune Privilege in Alopecia Areata

around affected anagen hair bulbs, is more attractive than even be transplanted into other tissues to create an immune
glucocorticoids. FK506 not only is a potent T- and NK-cell rejection-preventive milieu (Wang et al., 2015).
immunosuppressant and promotes regulatory T cell activ- Several immunocyte populations are now known to
ities (Arroyo Hornero et al., 2017; Shoughy, 2017), but it also actively participate in hair growth regulation, namely regu-
down-regulates MHC class Ia/b2 microglobulin expression in latory T cells (Ali et al., 2017; Bae et al., 2017; Castela et al.,
human anagen hair bulbs, protects human HFs from IFN- 2014; Conteduca et al., 2014; Han et al., 2015; Petukhova
geinduced IP collapse, and can even restore the latter et al., 2010), gd T-cells (Kloepper et al., 2013), macro-
ex vivo (Ito et al., 2004). Moreover, FK506 inhibits catagen phages (Castellana et al., 2014), and mast cells (Bertolini
development and promotes anagen in mice (Maurer et al, et al., 2014; Maurer et al., 1997a; Paus et al., 1994c;
1997b; Paus et al., 1996). A recent case report further en- Zhang et al., 2015). One important open question is
courages pursuit of this avenue of clinical investigation whether and how these immunocytes also affect HF IP. For
(Kanameishi et al., 2016). example, perifollicular mast cells may help to maintain
physiological HF IP (Bertolini et al., 2014) but may switch to
RECENT DEVELOPMENTS AND MAJOR OPEN a proinflammatory phenotype in AA that could both activate
QUESTIONS IN HF IP RESEARCH pathogenic CD8þ T cells and present autoantigens to them
The IP hair bulb epithelium must avoid constant attack by NK (Stelekati et al., 2009), thus contributing to triggering HF IP
cells, which recognize and eliminate MHC class Ia-negative collapse in AA. Indeed, antihistamines can reduce T-cell
cells. Therefore, one key HF IP characteristic is that chemotaxis in AA patients (Ito et al., 2013a). Furthermore,
NKG2D-activating ligands, such as MICA or ULBP3 (Elsner the role of resident, perifollicular memory T cells (Nasr et al.,
et al., 2010), are expressed at low levels in healthy HFs, 2005) and innate lymphoid cells (Klose and Artis, 2016) in
while these are greatly up-regulated in lesional AA HFs (Ito HF IP maintenance and collapse remains an important, as yet
et al., 2008a; Ito et al., 2008b; Petukhova et al., 2010; Xing unexplored AA research frontier.
et al., 2014). Recent evidence supports that overexpression Another crucial, but to our knowledge entirely unexplored
of both MICA and ULBP3 plays a role in AA pathobiology in research area, is how the microbiome of human HFs
different patient cohorts (Li et al., 2016; Moftah et al., 2016) (Scharschmidt et al., 2017) affects the HF IP. For example,
and therefore greatly increases the risk of HF IP collapse (Ito interindividual differences in the composition of the HF
et al., 2008a; Ito et al., 2008b). Most recently, we have found microbiome may modulate the balance between chemokine
that stressed human scalp HFs that overexpress MICA also and IP guardian secretion by HF keratinocytes and therefore
attract, activate, and induce IFN-g release from dermal play a significant role in determining the personal risk of
Vd1þT cells, leading to AA-like HF dystrophy and premature developing AA and response to therapy.
catagen induction ex vivo (Uchida et al., 2016). Finally, it has recently been hypothesized that HF IP may
Therefore, once the HF IP fails to avoid the stimulation of have been established as a mechanism to promote the de
NKG2Dþ immunocytes, massive influx of IFN-gesecreting novo induction of peripheral tolerance to neo-antigens
T cells may suffice to trigger the AA damage response occurring outside of the thymus that have thus escaped
pattern—perhaps even in the absence of antigen-dependent central tolerance (Oelert et al., 2016). If confirmed, proper
autoimmunity. Thus, it has become a central AA research functioning of the HF IP would be of fundamental importance
challenge to search for pharmacological strategies to down- for the maintenance of self-tolerance.
regulate the HF expression of activating NKG2D ligands
and to inhibit excessive activities of perifollicular NKG2Dþ IP-CENTERED FUTURE AA MANAGEMENT
cells (Ito et al., 2008a; Xing et al., 2014). Although therapy with JAK/STAT inhibitors has become a
If the location of the AA inflammatory infiltrate is linked to booming field of translational AA research (Jabbari et al.,
the presence of melanogenesis-associated (auto-)antigens in 2015; Kennedy Crispin et al., 2016; Mackay-Wiggan et al.,
the hair bulb (Paus et al., 1993), it is important to understand 2016), this must not detract from exploring other promising
whether interindividual differences in the robustness of the agents for AA management, namely candidate HF IP pro-
bulb IP (e.g., different constitutive expression levels of MHC tectants and restorers, which present an attractive cost-
class Ia/b2 microglobulin, “IP guardian,” MICA, and che- benefit-risk ratio.
mokine and chemokine receptors [Dai et al., 2016; Ito et al., Besides synthetic a-MSH analogs and FK506, these include
2013b, 2013c; Nagao et al., 2012], and intrafollicular ac- Kv1.3 channel inhibitors (Gilhar et al., 2013), PDE4
tivity of AIRE-regulated signaling [Kumar et al., 2011; inhibitors (Keren et al., 2015), substance P receptor antago-
Wengraf et al., 2008]) determine one’s personal risk of nists (Lindström et al., 2007), and VIP receptor (Bertolini
developing the AA response pattern once HF IP comes under et al., 2016) and CGRP receptor agonists (Kinori et al.,
threat, for example, after skin or HF trauma, viral infection, or 2012). Mono- or combination therapy with these agents
psychoemotional stress. may enhance the efficacy of HF IP restoration, thus sup-
Another important open question is whether and how the porting hair regrowth, and/or may protect the HF IP from
HF mesenchyme is involved in HF IP, besides harboring collapse, thus blocking AA progression.
immunoinhibitory immunocytes like mast cells that may More systematic research is also needed to clarify whether
contribute to the maintenance of HF IP (Bertolini et al., contact sensitizers (Lamb et al., 2016) restore HF IP, given
2014). Although the HF mesenchyme is clearly MHC class that hair regrowth after diphenylcyclopropenone therapy is
Ia/b2 microglobulin-positive, allotransplants of human HF associated with increased expression of the HF IP guardian
mesenchyme are not rejected (Reynolds et al., 1999) and can IL-10 (Hoffmann et al., 1994; Ito et al., 2004) and an altered

S14 Journal of Investigative Dermatology Symposium Proceedings (2018), Volume 19


R Paus et al.
Immune Privilege in Alopecia Areata

number of peribulbar CD8þ T cells, possibly indicating a Alli R, Nguyen P, Boyd K, Sundberg JP, Geiger TL. A mouse model of clonal
CD8þ T lymphocyte-mediated alopecia areata progressing to alopecia
change in CD8þ T-cell reactivity/function (Wasyłyszyn et al.,
universalis. J Immunol 2012;188:477e86.
2007). Could contact sensitizer therapy in AA be replaced
Alves de Medeiros AK, Speeckaert R, Desmet E, Van Gele M, De Schepper S,
with some of the agents described earlier to achieve more Lambert J1. JAK3 as an emerging target for topical treatment of inflam-
effective HF IP restoration and longer-lasting hair regrowth? matory skin diseases. PLoS One 2016;11:e0164080.
Finally, because low-dose IL-2 treatment has been report- Arroyo Hornero R, Betts GJ, Sawitzki B, Vogt K, Harden PN, Wood KJ.
edly beneficial in a small cohort of recalcitrant AA patients, CD45RA distinguishes CD4þCD25þCD127-/low TSDR demethylated
regulatory T cell subpopulations with differential stability and susceptibility
along with an increase in the number of regulatory T cells in
to tacrolimus-mediated inhibition of suppression. Transplantation
AA lesions (Castela et al., 2014), it will be interesting to 2017;101:302e9.
investigate how this IL-2 therapy affects HF IP. Bae JH, Hwang WS, Jang YJ, Lee YH, Jang DE, Kim JS, et al. CD80CD86
To establish a truly curative AA therapy, identification of deficiency disrupts regulatory CD4þFoxP3þT cell homeostasis and
the autoantigens in AA (more precisely, in AAA [see above]) induces autoimmune-like alopecia [e-pub ahead of print]. Exp Dermatol
and of the T-cell receptor clonotypes of autoreactive CD8þ T- 2017;26:1053e9.
Bertolini M, Pretzlaff M, Sulk M, Bähr M, Gherardini J, Uchida Y, et al.
cells is required, at least for the subgroup of patients for
Vasoactive intestinal peptide, whose receptor-mediated signalling may be
whom the AA response pattern reflects an antigen-specific defective in alopecia areata, provides protection from hair follicle immune
autoimmune response. The ongoing attempts to identify privilege collapse. Br J Dermatol 2016;175:531e41.
either (e.g., Bertolini et al., 2015, 2017; Gilhar et al., 2001; Bertolini M, Rossi A, Paus R. Cover Image: Are melanocyte-associated pep-
Ito et al., 2014; Wang et al., 2016) are of the utmost impor- tides the elusive autoantigens in alopecia areata? Br J Dermatol 2017;176:
1106.
tance. However, if the hypothesis is correct that AA is just a
Bertolini M, Uchida Y, Paus R. Toward the clonotype analysis of alopecia
stereotypic response pattern of anagen HFs to excessive
areata-specific, intralesional human CD8þ T lymphocytes. J Investig Der-
proinflammatory signals triggered by several distinct patho- matol Symp Proc 2015;17:9e12.
mechanisms, of which autoantigen- and CD8þ T- Bertolini M, Zilio F, Rossi A, Kleditzsch P, Emelianov VE, Gilhar A, et al.
celledependent autoimmunity is only one among others Abnormal interactions between perifollicular mast cells and CD8þ T-cells
(Paus, in: McElwee et al., 2013), one must conclude that may contribute to the pathogenesis of alopecia areata. PLoS One 2014;9:
e94260.
curative AA therapy based on tolerization against identified
Betz RC, Petukhova L, Ripke S, Huang H, Menelaou A, Redler S, et al.
AA autoantigens or elimination of autoreactive T cells is
Genome-wide meta-analysis in alopecia areata resolves HLA associations
attainable for only some patients: those in whom genuine and reveals two new susceptibility loci. Nat Commun 2015;6:5966.
anti-HF autoimmunity underlies the AA hair loss phenotype Billingham RE, Silvers WK. A biologist’s reflections on dermatology. J Invest
(AAA). Dermatol 1971;57:227e40.
In contrast, all AA patients will benefit from therapy that Breitkopf T, Lo BK, Leung G, Wang E, Yu M, Carr N, et al. Somatostatin
manages to rapidly and lastingly restore HF IP, ideally at expression in human hair follicles and its potential role in immune privi-
lege. J Invest Dermatol 2013;133:1722e30.
minimal risk of adverse effects. Therefore, investigators
interested in developing more effective AA therapy are Brzoska T, Luger TA, Maaser C, Abels C, Böhm M. Alpha-melanocyte-
stimulating hormone and related tripeptides: biochemistry, antiin-
encouraged to turn their full attention to understanding the flammatory and protective effects in vitro and in vivo, and future
mechanisms that jointly maintain HF IP, while patient advo- perspectives for the treatment of immune-mediated inflammatory diseases.
cacy groups such as NAAF (www.naaf.org) should persuade Endocr Rev 2008;29:581e602.
policy makers to invest more funding into HF IP research. Castela E, Le Duff F, Butori C, Ticchioni M, Hofman P, Bahadoran P, et al.
Effects of low-dose recombinant interleukin 2 to promote T-regulatory cells
Beyond AA, other patients with IP collapse-related autoim-
in alopecia areata. JAMA Dermatol 2014;150:748e51.
mune diseases are likely to benefit from having these ques-
Castellana D, Paus R, Perez-Moreno M. Macrophages contribute to the cyclic
tions answered in this model disease. activation of adult hair follicle stem cells. PLoS Biol 2014;12:e1002002.
CONFLICT OF INTEREST Conteduca G, Rossi A, Megiorni F, Parodi A, Ferrera F, Tardito S, et al. Single
The authors state no conflict of interest. nucleotide polymorphisms in the promoter regions of Foxp3 and ICOSLG
genes are associated with Alopecia areata. Clin Exp Med 2014;14:91e7.
Chang SH, Jung EJ, Lim DG, Park YH, Wee YM, Kim JH, et al. Anti-
ACKNOWLEDGMENTS inflammatory action of alpha-melanocyte stimulating hormone (alpha-
Writing of this review was made possible by grants from NAAF (USA) to RP MSH) in anti-CD3/CD28-mediated spleen and CD4(þ)CD25(-) T cells and
and MB, from National Institute of Health Research Manchester Biomedical a partial participation of IL-10. Immunol Lett 2008;118:44e8.
Research Centre (Inflammatory Hair Diseases Programme) to RP and SBP, and Christoph T, Müller-Röver S, Audring H, Tobin DJ, Hermes B, Cotsarelis G,
from Associazione Mediterranea Alopecia Areata (Italy) to MB. et al. The human hair follicle immune system: cellular composition and
Funding for the Summit and the publication of this article was provided by immune privilege. Br J Dermatol 2000;142:862e73.
the National Alopecia Areata Foundation. Funding for this Summit was also
made possible (in part) by a grant (1 R13AR071266) from the National Dai Z, Xing L, Cerise J, Wang EH, Jabbari A, de Jong A, et al. CXCR3 blockade
Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS). The inhibits T cell migration into the skin and prevents development of alopecia
views expressed in written conference materials or publications and by areata. J Immunol 2016;197:1089e99.
speakers and moderators do not necessarily reflect the official policies of the Elsner L, Flügge PF, Lozano J, Muppala V, Eiz-Vesper B, Demiroglu SY, et al.
Department of Health and Human Services; nor does mention of trade names, The endogenous danger signals HSP70 and MICA cooperate in the acti-
commercial practices, or organizations imply endorsement by the U.S. vation of cytotoxic effector functions of NK cells. J Cell Mol Med 2010;14:
Government. 992e1002.
Eleftheriadis T, Pissas G, Antoniadi G, Liakopoulos V, Tsogka K, Sounidaki M,
et al. Differential effects of the two amino acid sensing systems, the GCN2
REFERENCES kinase and the mTOR complex 1, on primary human alloreactive CD4⁺
Ali N, Zirak B, Rodriguez RS, Pauli ML, Truong HA, Lai K, et al. Regulatory T-cells. Int J Mol Med 2016;37:1412e20.
T cells in skin facilitate epithelial stem cell differentiation. Cell 2017;169: Engelhardt B, Vajkoczy P, Weller RO. The movers and shapers in immune
1119e29. privilege of the CNS. Nat Immunol 2017;18:123e31.

www.jidsponline.org S15
R Paus et al.
Immune Privilege in Alopecia Areata

Fischer J, Degenhardt F, Hofmann A, Redler S, Basmanav FB, Heilmann- Jabbari A, Dai Z, Xing L, Cerise JE, Ramot Y, Berkun Y, et al. Reversal of
Heimbach S, et al. Genome-wide analysis of copy number variants in alopecia areata following treatment with the JAK1/2 inhibitor baricitinib.
alopecia areata in a Central European cohort reveals association with EBioMedicine 2015;2:351e5.
MCHR2. Exp Dermatol 2016;26:536e41. Joyce JA, Fearon DT. T cell exclusion, immune privilege, and the tumor
Gao Z, Jin YQ, Wu W. SOCS3 treatment prevents the development of alo- microenvironment. Science 2015;348:74e80.
pecia areata by inhibiting CD8þ T cell-mediated autoimmune destruction. Kanameishi S, Dainichi T, Endo Y, Otsuka A, Tanioka M, Kabashima K.
Oncotarget 2017;8:33432e43. Successful hair regrowth in an acute diffuse form of alopecia areata during
Giangreco A, Hoste E, Takai Y, Rosewell I, Watt FM. Epidermal Cadm1 oral tacrolimus treatment in a patient with rheumatoid arthritis. J Eur Acad
expression promotes autoimmune alopecia via enhanced T cell adhesion Dermatol Venereol 2016;31:3137e8.
and cytotoxicity. J Immunol 2012;188:1514e22. Kang H, Wu WY, Lo BK, Yu M, Leung G, Shapiro J, et al. Hair follicles from
Gilhar A, Etzioni A, Paus R. Alopecia areata. N Engl J Med 2012;366: alopecia areata patients exhibit alterations in immune privilege-associated
1515e25. gene expression in advance of hair loss. J Invest Dermatol 2010;130:
Gilhar A, Keren A, Shemer A, Ullmann Y, Paus R. Blocking potassium 2677e80.
channels (Kv1.3): a new treatment option for alopecia areata? J Invest Kassira S, Korta DZ, W Chapman L, Dann F. Review of treatment for alopecia
Dermatol 2013;133:2088e91. totalis and alopecia universalis. Int J Dermatol 2017;56:801e10.
Gilhar A, Landau M, Assy B, Ullmann Y, Shalaginov R, Serafimovich S, et al. Kennedy Crispin M, Ko JM, Craiglow BG, Li S, Shankar G, Urban JR, et al.
Melanocyte-associated T cell epitopes can function as autoantigens for Safety and efficacy of the JAK inhibitor tofacitinib citrate in patients with
transfer of alopecia areata to human scalp explants on Prkdc(scid) mice. alopecia areata. JCI Insight 2016;1:e89776.
J Invest Dermatol 2001;117:1357e62. Keren A, Shemer A, Ullmann Y, Paus R, Gilhar A. The PDE4 inhibitor, apre-
Gilhar A, Schrum AG, Etzioni A, Waldmann H, Paus R. Alopecia areata: milast, suppresses experimentally induced alopecia areata in human skin
Animal models illuminate autoimmune pathogenesis and novel immuno- in vivo. J Dermatol Sci 2015;77:74e6.
therapeutic strategies. Autoimmun Rev 2016;15:726e35. Kinori M, Bertolini M, Funk W, Samuelov L, Meyer KC, Emelianov VU, et al.
Gilhar A, Ullmann Y, Berkutzki T, Assy B, Kalish RS. Autoimmune hair loss Calcitonin gene-related peptide (CGRP) may award relative protection
(alopecia areata) transferred by T lymphocytes to human scalp explants on from interferon-g-induced collapse of human hair follicle immune privi-
SCID mice. J Clin Invest 1998;101:62e7. lege. Exp Dermatol 2012;21:223e6.
Han YM, Sheng YY, Xu F, Qi SS, Liu XJ, Hu RM, et al. Imbalance of T-helper Kinori M, Kloepper JE, Paus R. Can the hair follicle become a model for
17 and regulatory T cells in patients with alopecia areata. J Dermatol studying selected aspects of human ocular immune privilege? Invest
2015;42:981e8. Ophthalmol Vis Sci 2011;52:4447e58.
Harel S, Higgins CA, Cerise JE, Dai Z, Chen JC, et al. Pharmacologic inhi- Kloepper JE, Kawai K, Bertolini M, Kanekura T, Paus R. Loss of gd T cells
bition of JAK-STAT signaling promotes hair growth. Sci Adv 2015;1: results in hair cycling defects. J Invest Dermatol 2013;133:1666e9.
e1500973. Klose CS, Artis D. Innate lymphoid cells as regulators of immunity, inflam-
Harrist TJ, Ruiter DJ, Mihm MC Jr, Bhan AK. Distribution of major mation and tissuehomeostasis. Nat Immunol 2016;17:765e74.
histocompatibility antigens in normal skin. Br J Dermatol 1983;109: Kostovic K, Gulin SJ, Mokos ZB, Ceovic R. Tofacitinib, an oral janus
623e33. kinase inhibitor: perspectives in dermatology. Curr Med Chem 2017;24:
Hoffman U, Tokura Y, Nishijima T, Takigawa M, Paus R. Hair cycle-dependent 1158e67.
changes in skin immune functions: anagen-associated depression of Kumar V, Pedroza LA, Mace EM, Seeholzer S, Cotsarelis G, Condino-Neto A,
sensitization for contact hypersensitivity in mice. J Invest Dermatol et al. The autoimmune regulator (AIRE), which is defective in autoimmune
1996;106:598e604. polyendocrinopathy-candidiasis-ectodermal dystrophy patients, is
Hoffmann R, Wenzel E, Huth A, van der Steen P, Schäufele M, Henninger HP, expressed in human epidermal and follicular keratinocytes and associates
et al. Cytokine mRNA levels in Alopecia areata before and after treatment with the intermediate filament protein cytokeratin 17. Am J Pathol
with the contact allergen diphenylcyclopropenone. J Invest Dermatol 2011;178:983e8.
1994;103:530e3. Lamb RC, Young D, Holmes S. Retrospective review of diphencyprone in the
Ikeda T. A new classification of alopecia areata. Dermatologica 1965;131: treatment of alopecia areata. Clin Exp Dermatol 2016;41:352e8.
421e45. Li J, van Vliet C, Rufaut NW, Jones LN, Sinclair RD, Carbone FR. Laser capture
Ito T, Bertolini M, Funakoshi A, Ito N, Takayama T, Biro T, Paus R, et al. Birth, microdissection reveals transcriptional abnormalities in alopecia areata
life, and death of the MAGE3 hypothesis of alopecia areata pathobiology. before, during, and after active hair loss. J Invest Dermatol 2016;136:
J Dermatol Sci 2013a;72:327e30. 715e8.
Ito T, Fujiyama T, Hashizume H, Tokura Y. Antihistaminic drug olopatadine Lindström E, von Mentzer B, Påhlman I, Ahlstedt I, Uvebrant A, Kristensson E,
downmodulates T cell chemotaxis toward CXCL10 by reducing CXCR3 et al. Neurokinin 1 receptor antagonists: correlation between in vitro re-
expression, F-actin polymerization and calcium influx in patients with ceptor interaction and in vivo efficacy. J Pharmacol Exp Ther 2007;322:
alopecia areata. J Dermatol Sci 2013b;72:68e71. 1286e93.
Ito T, Hashizume H, Shimauchi T, Funakoshi A, Ito N, Fukamizu H, et al. Liu LY, Craiglow BG, Dai F, King BA. Tofacitinib for the treatment of severe
CXCL10 produced from hair follicles induces Th1 and Tc1 cell infiltration alopecia areata and variants: a study of 90 patients. J Am Acad Dermatol
in the acute phase of alopecia areata followed by sustained Tc1 accumu- 2017;76:22e8.
lation in the chronic phase. J Dermatol Sci 2013c;69:140e7. Ma EH, Bantug G, Griss T, Condotta S, Johnson RM, Samborska B, et al.
Ito T, Ito N, Bettermann A, Tokura Y, Takigawa M, Paus R. Collapse and Serine is an essential metabolite for effector T cell expansion. Cell Metab
restoration of MHC class-I-dependent immune privilege: exploiting the 2017;25:345e57.
human hair follicle as a model. Am J Pathol 2004;164:623e34. Mackay-Wiggan J, Jabbari A, Nguyen N, Cerise JE, Clark C, Ulerio G, et al.
Ito T, Ito N, Saathoff M, Bettermann A, Takigawa M, Paus R. Interferon-gamma Oral ruxolitinib induces hair regrowth in patients with moderate-to-severe
is a potent inducer of catagen-like changes in cultured human anagen hair alopecia areata. JCI Insight 2016;1:e89790.
follicles. Br J Dermatol 2005;152:623e31. Maurer M, Fischer E, Handjiski B, von Stebut E, Algermissen B, Bavandi A,
Ito T, Ito N, Saatoff M, Hashizume H, Fukamizu H, Nickoloff BJ, et al. et al. Activated skin mast cells are involved in murine hair follicle regres-
Maintenance of hair follicle immune privilege is linked to prevention of NK sion (catagen). Lab Invest 1997a;77:319e32.
cell attack. J Invest Dermatol 2008a;128:1196e206. Maurer M, Handjiski B, Paus R. Hair growth modulation by topical immu-
Ito T, Meyer KC, Ito N, Paus R. Immune privilege and the skin. Curr Dir nophilin ligands: induction of anagen, inhibition of massive catagen
Autoimmun 2008b;10:27e52. development, and relative protection from chemotherapy-induced alope-
Ito T, Tokura Y. The role of cytokines and chemokines in the T-cell- cia. Am J Pathol 1997b;150:1433e41.
mediated autoimmune process in alopecia areata. Exp Dermatol McElwee KJ, Gilhar A, Tobin DJ, Ramot Y, Sundberg JP, Nakamura M, et al.
2014;23:787e91. What causes alopecia areata? Exp Dermatol 2013;22:609e26.

S16 Journal of Investigative Dermatology Symposium Proceedings (2018), Volume 19


R Paus et al.
Immune Privilege in Alopecia Areata

Meyer KC, Klatte JE, Dinh HV, Harries MJ, Reithmayer K, Meyer W, et al. Reynolds AJ, Lawrence C, Cserhalmi-Friedman PB, Christiano AM,
Evidence that the bulge region is a site of relative immune privilege in Jahoda CA. Trans-gender induction of hair follicles. Nature 1999;402:
human hair follicles. Br J Dermatol 2008;159:1077e85. 33e4.
Medawar PB. Tests by tissue culture methods on the nature of immunity to Scharschmidt TC, Vasquez KS, Pauli ML, Leitner EG, Chu K, Truong HA, et al.
transplanted skin. Q J Microsc Sci 1948;89:239e52. Commensal microbes and hair follicle morphogenesis coordinately drive
Miteva M, Misciali C, Fanti PA, Tosti A. Histopathologic features of alopecia Treg migration into neonatal skin. Cell Host Microbe 2017;21:467e477.
areata incognito: a review of 46 cases. J Cutan Pathol 2012;39:596e602. e5.

Moftah NH, El-Barbary RA, Rashed L, Said M. ULBP3: a marker for alopecia Shoughy SS. Topical tacrolimus in anterior segment inflammatory disorders.
areata incognita. Arch Dermatol Res 2016;308:415e21. Eye Vis 2017;4:7.

Nagao K, Kobayashi T, Moro K, Ohyama M, Adachi T, Kitashima DY, et al. Stelekati E, Bahri R, D’Orlando O, Orinska Z, Mittrücker HW, Langenhaun R,
Stress-induced production of chemokines by hair follicles regulates the et al. Mast cell-mediated antigen presentation regulates CD8þ T cell
trafficking of dendritic cells in skin. Nat Immunol 2012;13:744e52. effector functions. Immunity 2009;31:665e76.

Nasr IW, Wang Y, Gao G, Deng S, Diggs L, Rothstein DM, et al. Testicular Tan PH, Bharath AK. Manipulation of indoleamine 2,3 dioxygenase; a novel
immune privilege promotes transplantation tolerance by altering the bal- therapeutic target for treatment of diseases. Expert Opin Ther Targets
ance between memory and regulatory T cells. J Immunol 2005;174: 2009;13:987e1012.
6161e8. Taylor AW. Ocular immune privilege and transplantation. Front Immunol
Noso S, Park C, Babaya N, Hiromine Y, Harada T, Ito H, et al. Organ speci- 2016;7:37.
ficity in autoimmune diseases: thyroid and islet autoimmunity in alopecia Tosti A, Bellavista S, Iorizzo M. Alopecia areata: a long term follow-up study
areata. J Clin Endocrinol Metab 2015;100:1976e83. of 191 patients. J Am Acad Dermatol 2006;55:438e41.
Oelert T, Gilhar A, Paus R. T cell “induced-self” MHC class I/peptide com- Truckenmiller ME, Princiotta MF, Norbury CC, Bonneau RH. Corticosterone
plexes may enable “de novo” tolerance induction to neo-antigens occur- impairs MHC class I antigen presentation by dendritic cells via reduction of
ring outside of the thymus. Exp Dermatol 2016;26:529e31. peptide generation. J Neuroimmunol 2005;160:48e60.
Oh JW, Kloepper J, Langan EA, Kim Y, Yeo J, Kim MJ, et al. A guide to studying Uchida Y, Gherardini J, Alam M, Keren A, Arakawa A, Rossi A, et al.
human hair follicle cycling in vivo. J Invest Dermatol 2016;136:34e44. Vd1þT-cells are stress-sentinels in human skin and are implicated in alo-
Paus R, Böttge JA, Henz BM, Maurer M. Hair growth control by immuno- pecia areata pathogenesis. J Invest Dermatol 2016;136(9 Suppl. 2):S228.
suppression. Arch Dermatol Res 1996;288:408e10. Wang EH, Yu M, Breitkopf T, Akhoundsadegh N, Wang X, Shi FT, et al.
Paus R, Eichmüller S, Hofmann U, Czarnetzki BM, Robinson P. Expression of Identification of autoantigen epitopes in alopecia areata. J Invest Dermatol
classical and non-classical MHC class I antigens in murine hair follicles. Br 2016;136:1617e26.
J Dermatol 1994a;131:177e83. Wang X, Marr AK, Breitkopf T, Leung G, Hao J, Wang E, et al. Hair follicle
Paus R, Hofmann U, Eichmüller S, Czarnetzki BM. Distribution and changing mesenchyme-associated PD-L1 regulates T-cell activation induced
density of gamma-delta T cells in murine skin during the induced hair apoptosis: a potential mechanism of immune privilege. J Invest Dermatol
cycle. Br J Dermatol 1994b;130:281e9. 2014;134:736e45.
Paus R, Maurer M, Slominski A, Czarnetzki BM. Mast cell involvement in Wang X, Hao J, Leung G, Breitkopf T, Wang E, Kwong N, et al. Hair follicle
murine hair growth. Dev Biol 1994c;163:230e40. dermal sheath derived cells improve islet allograft survival without sys-
temic immunosuppression. J Immunol Res 2015;2015:607328.
Paus R, Nickoloff BJ, Ito T. A ‘hairy’ privilege. Trends Immunol 2005;26:
Wasyłyszyn T, Kozłowski W, Zabielski SL. Changes in distribution pattern of
32e40.
CD8 lymphocytes in the scalp in alopecia areata during treatment with
Paus R, Slominski A, Czarnetzki BM. Is alopecia areata an autoimmune- diphencyprone. Arch Dermatol Res 2007;299:231e7.
response against melanogenesis-related proteins, exposed by abnormal
Wengraf DA, McDonagh AJ, Lovewell TR, Vasilopoulos Y, Macdonald-
MHC class I expression in the anagen hair bulb? Yale J Biol Med 1993;66:
Hull SP, Cork MJ, et al. Genetic analysis of autoimmune regulator haplo-
541e54.
types in alopecia areata. Tissue Antigens 2008;71:206e12.
Paus R, van der Veen C, Eichmüller S, Kopp T, Hagen E, Müller-Röver S, et al. Westgate GE, Craggs RI, Gibson WT. Immune privilege in hair growth. J Invest
Generation and cyclic remodeling of the hair follicle immune system in Dermatol 1991;97:417e20.
mice. J Invest Dermatol 1998;111:7e18.
Xing L, Dai Z, Jabbari A, Cerise JE, Higgins CA, Gong W, et al. Alopecia areata
Peters EMJ, Liotiri S, Bodó E, Hagen E, Bı́ró T, Arck PC, Paus R. Probing the is driven by cytotoxic T lymphocytes and is reversed by JAK inhibition. Nat
effects of stress mediators on the human hair follicle: substance P holds Med 2014;20:1043e9.
central position. Am J Pathol 2007;171:1872e86.
Zhang X, Zhao Y, Ye Y, Li S, Qi S, Yang Y, et al. Lesional infiltration of mast
Petukhova L, Duvic M, Hordinsky M, Norris D, Price V, Shimomura Y, et al. cells, Langerhans cells, T cells and local cytokine profiles in alopecia
Genome-wide association study in alopecia areata implicates both innate areata. Arch Dermatol Res 2015;307:319e31.
and adaptive immunity. Nature 2010;466:113e7.
Zarbo A, Belum VR, Sibaud V, Oudard S, Postow MA, Hsieh JJ, et al. Immune-
Pratt CH, King LE Jr, Messenger AG, Christiano AM, Sundberg JP. Alopecia related alopecia (areata and universalis) in cancer patients receiving
areata. Nat Rev Dis Primers 2017;16(3):17011. immune checkpoint inhibitors. Br J Dermatol 2017;176:1649e52.

www.jidsponline.org S17

You might also like