You are on page 1of 16

Semin Immunopathol (2013) 35:677–691

DOI 10.1007/s00281-013-0394-4

REVIEW

Functions of the aryl hydrocarbon receptor in the skin


Charlotte Esser & Imke Bargen & Heike Weighardt &
Thomas Haarmann-Stemmann & Jean Krutmann

Received: 21 January 2013 / Accepted: 16 July 2013 / Published online: 16 August 2013
# Springer-Verlag Berlin Heidelberg 2013

Abstract Among other functions, the skin serves as the Background


barrier against the environment and provides vital pro-
tection from physical or chemical harm and from in- The skin covers the whole body and is in constant contact with
fection. Skin cells express the aryl hydrocarbon recep- environmental factors, such as heat and cold, sunlight, any
tor (AHR), a ligand-activated transcription factor and chemicals one touches or applies, bacteria, viruses, etc. The
sensor of environmental chemicals; at the same time, skin is uniquely suited to this task, and forms an effective
AHR ligands are abundant in skin from exogenous or barrier against environmental harm, while protecting the in-
endogenous sources. For example, solar radiation, in tegrity of the body.
particular ultraviolet (UV) B, generates AHR ligands Integration of endogenous and exogenous signals is a
from tryptophan in the skin. Recent evidence has challenge for all cells and organs. Receptors, which can sense
shown that AHR is involved in the (patho)physiology environmental signals and physiological/metabolic imbal-
of skin including the regulation of skin pigmentation, ances, trigger intracellular events cumulating in transcription,
photocarcinogenesis, and skin inflammation. We here protein expression, and cellular change. The areas of the body,
provide a state-of-the-art summary of work which re- which are in direct contact with the environment, are endowed
lates to the role of the AHR in (1) adaptive responses with special cells and mechanisms for the response to envi-
against environmental challenges such as UVB or top- ronmental situations. A major sensor of chemical signals is the
ical chemicals and (2) intrinsic developmental roles for aryl hydrocarbon receptor (AHR), a member of the evolution-
homeostasis of skin cells and (3) skin immunity. We arily old family of per-arnt-sim basic helix-loop-helix tran-
also discuss the existing evidence that AHR antagonists scription factors (PAS-bHLH). Similar to the steroid and
or AHR ligands may be used for the prevention and/or glucocorticoid hormones, AHR resides in the cytoplasm and
treatment of skin disease. becomes activated by certain low molecular weight chemicals.
Both murine and human AHR have been cloned and the basic
biochemistry has been worked out many years ago [1, 2].
Keywords Aryl hydrocarbon receptor . Epidermis . Briefly, AHR sheds its chaperoning proteins (hsp90, c-src,
Langerhans cells . Psoriasis . Skin cancer . Pigmentation . AIP, and others) upon ligand binding, which exposes a nuclear
FICZ . UV translocation signal. AHR moves into the nucleus and dimer-
izes with AHR nuclear transporter (ARNT), another member
of the PAS-bHLH family. The complex induces transcription
of genes possessing conserved AHR binding sites (called
xenobiotic response elements, XREs) in their promoters.
This article is a contribution to the special issue on Roles of Aryl Eventually, AHR is exported again into the cytosol and de-
Hydrocarbon Receptor in Controlling Immunity - Guest Editors: C. Pot, graded [1, 3]. Many genes contain functional XREs, and AHR
V. Kuchroo, and F. Quintaña
regulates a plethora of genes in a cell-, tissue-, and condition-
C. Esser (*) : I. Bargen : H. Weighardt : T. Haarmann-Stemmann : specific fashion [4, 5]. The quality and duration of AHR
J. Krutmann
activation by various ligands directs level and spectrum of
Leibniz-Research Institute for Environmental Medicine (IUF),
Auf’m Hennekamp 50, 40225 Dusseldorf, Germany the genes which are induced, and are thus pivotal in the
e-mail: chesser@uni-duesseldorf.de outcome, including a “toxic” outcome [6]. Three important
678 Semin Immunopathol (2013) 35:677–691

groups of genes are targeted by AHR. First, the battery of genes innate and adaptive immune responses, metabolizes and ex-
responsible for metabolism of ring molecules, such as cyto- cludes external chemicals, and protects itself against DNA
chrome P (CYP) gene cyp1a1 or nqo1; second, genes related damage by UV radiation via pigmentation and detection/
to development, cell cycle, and cell differentiation; and finally, elimination of cancer cells. Furthermore, the skin has the
genes related to immunity. Other highly cell-specific gene func- capacity to heal wounds. Keratinocyte stem cells are found at
tions can be found as well. It is increasingly appreciated that the dermal/epidermal interface. They differentiate progressive-
AHR can cross-feed into other signaling pathways, such as ly and vertically from this basal stratum, first into strata of
NFκB, allowing for a highly differentiated cellular response metabolically active spinous and granular cells, which produce
[7]. Moreover, ligand activation dissociates the protein tyrosine keratin and lipids. Eventually, in the stratum corneum,
kinase pp60src from the cytosolic AHR complex, which can keratinocytes lose their nucleus and form a so-called cornified
move to the cell membrane-bound epidermal growth factor envelope, a rigid structure composed of lipids enriched in
receptor (EGFR), and thus trigger downstream MAPKs [8]. ceramides and cholesterol. The entire process renews the skin
The AHR pathway is downregulated by the AHR repressor continuously over lifetime. Interspersed and in direct contact
(AHRR), a target gene of the AHR in a negative feedback loop. with the keratinocytes are melanocytes (MCs) (approximately
The AHRR competes with AHR for the binding site of ARNT 5 %), Langerhans cells (LCs) (1–3 %), the dendritic cells
and forms a transcriptionally inactive complex [9]. It may, more- (DCs) of the skin, αβ T cells, and T cells with an invariant
over, have functions beyond simple repression of AHR [10]. γδ T cell receptor (1–5 %). The latter are often referred to as
Expression levels of AHR are especially high in the liver dendritic epidermal T cells due to their shape. Every LC and
and in the barrier organs, such as lung, gut, and skin. This fits MC is in contact with many keratinocytes, forming a tight
well with both the traditionally well-known function of AHR network. In addition, mast cells are located just underneath
in xenobiotic metabolism and with its newly discovered roles the epidermis. The dermis—with fibroblasts as the structure
as a major player in the immune response and cell homeosta- giving cell type—has an anatomically more complex structure
sis. Finally, AHR has a role in development of some organs and higher cell diversity, it harbors macrophages, mast cells,
and tissues. additional subsets of dendritic cells [11], and αβ T cells as
AHR is expressed in all skin cells, and endogenous ligands well, which vastly increase in numbers in the inflamed state.
are formed in situ from tryptophan via ultraviolet (UV) radia- For humans, it has been estimated that skin contains approxi-
tion. Interestingly, chloracne and aberrant pigmentation are two mately 1 million T cells/cm2, thus there are more T cells in the
major adverse effects of exposure to the high-affinity AHR skin than in the blood [12]. The immunological challenge of
ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a notori- the skin is the balance between protecting against a constant
ously toxic environmental pollutant. Epidemiological identified threat of pathogens while suppressing unnecessary and harm-
correlations between skin cancer and exposure to AHR ligands ful inflammation [13]. The skin is exposed to harmful or
in workers (such as coal miners) or by lifestyle (e.g., smokers) damaging high energy UV radiation from the sun. Repair of
are well known. Knowledge on the physiology of AHR in the DNA damage, removal of dead cells if necessary, and surveil-
skin increased tremendously over the last years and highlighted lance against cancer cells is therefore another vital task of the
that AHR is involved in many aspects of skin physiology, such skin. The AHR emerges as an important player in skin homeo-
as detoxification, cellular homeostasis, skin pigmentation, and stasis, as well as during infection and inflammation (Fig. 1).
skin immunity. Here, we review the current knowledge of the
expression and function of AHR in the skin, and discuss the
therapeutic potential of manipulating this signaling system. Expression of the aryl hydrocarbon receptor in skin cells

It had been noted already in the 1990s that AHR expression


The skin—a layered organ with many functions levels differed vastly between organs in both laboratory ani-
mals and in humans [2, 14, 15]. In these reports, liver and lung
The skin is a layered organ of tightly connected cells. It has were the organs with the highest AHR content, while muscle
three main layers, the subcutis, the dermis (mainly fibroblasts), and brain had very low levels. In the work from our institute,
and the thinner outer epidermis (with keratinocytes (KC) as the we data-mined almost 2,000 published gene expression pro-
construction cells). Located in the dermis are sensory organs files from various tissues and cell lines, and identified lineage
for temperature and touch, hair bulbs, sebaceous glands, nerve and differentiation specific changes in AHR expression in T
cells, blood vessels, and lymphatics, which serve as ports for cells and B cells [5]. More recently, various cell types, in
leukocyte migration in and out of the skin. The epidermis particular splenic B cells, T cells, and dendritic cells, were
serves critical functions in body protection. It is a physical/ sorted to high purity and analyzed for AHR mRNA or protein
mechanical, chemical, and biological barrier. It protects against expression [16]. With respect to the skin, most cell types of the
transcutaneous water loss, fights off microbes and viruses by epidermis and fibroblasts in the dermis have been analyzed by
Semin Immunopathol (2013) 35:677–691 679

Fig. 1 Schematic overview of


skin structure and AHR
expression in skin cells. The
various cell populations and their
situation in skin are indicated. The
intensity of gray shading
indicates AHR expression levels.
White cells not studied, hatched
cells both AHR and AHR-
repressor expression were
reported

now. It is noteworthy that they all, without exception, have indolcarbinols. These substances can reach the skin either via
high levels of AHR expression. The expression of AHR in the food chain or by direct contact or topical application (e.g., if
different cell types of the skin is summarized in Table 1. they are present in cosmetics or sunscreens). Even the skin
Albeit, it appears self-evident that constitutive expression of microflora can produce ligands. Finally, endogenous AHR li-
AHR is necessary for its action, no simple correlation exists gands are formed in the body, such as ITE, a tryptophan–cystein
between expression levels, ligand sensitivity, strength of bio- dimer [17, 18], kynurenines, or 6-formylindolo[3,2-b]carbazole
logical response, or biological importance of AHR for the cell (FICZ), a tryptophan dimer. Some place FICZ into the exoge-
[5]. Most likely, expression level is one facet in the multilay- nous group as it is generated under the exposure to UVB or solar
ered regulation of this complex signaling pathway. light, not via a metabolic pathway. Because different ligands
result in different outcomes [6, 17], it is generally assumed that
the nature of the ligand—availability, structure, half-life, etc.—
Natural and xenobiotic AHR ligands in the skin determines the AHR response [19]. There is still plenty of
are abundant unknown terrain, which needs to be covered in order to exploit
ligands therapeutically (Table 2).
AHR can bind to and become activated by sterically planar
ligands with an approximate size of three benzene rings [17].
Ligands can be grouped in three ways. First, exogenous/ FICZ generated by UV radiation
synthetic, such as the high-affinity pollutant TCDD and other
polycyclic aromatic hydrocarbons (PAH) (e.g., biphenyls, 7,12- Directly in the skin, solar radiation and especially UVB gen-
dimethylbenz[a]anthracene (DMBA), methylcholanthrene, or erate tryptophan dimers of the indolo[3,2-b]carbazole type.
benzo[a]pyrene (B[a]P)). A second group would be the The UV/light-generated ligands were chemically characterized
exogenous/natural compounds, which are found in or metabo- and identified as FICZ and 6,12-diformylindolo[3,2-b]carba-
lized from dietary plants such as glucosinolates, flavonoids, or zole [20], and others. First detected in vitro (7), generation of
680 Semin Immunopathol (2013) 35:677–691

Table 1 Expression of AHR in skin cells

Mouse Human

Cell type (main functions) Constitutive AHR Reference Cell type AHR Reference
expression expression

Keratinocytes (barrier function, elasticity, filter out UV Yes [33] HaCaT (keratinocyte cell line) Yes [8]
damage, produce cytokines)
Langerhans cellsa (take up antigen, produce cytokines) Yes [33] Stem cell derived LC Yes [127]
Vγ5 γδ T cellsa (immunosurveillance, wound healing) Yes [102] γδ T cells ND
Melanocytes (pigmentation) Yes [53] Cultured foreskin melanocytes Yes [55]
Mast cells Yes [128] Yes [128]
Fibroblastsa (give pliability) Yes [129] Fibroblastsb Yes [10]
Sebocytes ND Cultured SZ95 sebocytes Yes [130]
linnegSca1neg bone marrow progenitors (as a source for Yes [131]
in vitro generated LC)

The table does not give quantitative measurements because expression levels cannot be compared across the various published data and between cell
types. In some papers, expression levels were compared to liver (which has one of the highest expressions in body tissues). It might be said, though, that
expression levels in all skin cell subsets is generally high. Expression was measured by either RT-PCR, Western blotting, or immunohistochemistry.
Where done, there appeared to be a good correlation between PCR-data and protein data. AHR is not or only very weakly expressed in naïve T cells,
muscle tissues, brain, and spleen (both in human and in mouse) [2, 132]; ND no data available
a
High AHR-repressor expression was reported in these cells
b
High AHR-repressor was reported in foreskin-derived fibroblasts, but not in adult fibroblast from mammary skin

such ligands by UVB irradiation was confirmed in vivo in AHR may serve as a sensor for UV radiation, and its formation
hepatocytes and keratinocytes as well [8, 21–23]. They have explains cutaneous as well as extracutaneous induction of
very high affinities to AHR, similar to TCDD, but are rapidly CYP1A1 enzyme activity after skin exposure to UV radiation
degraded within cells. Simple solar light suffices to generate [8, 27]. It remains to be shown, if UV also enhances metabo-
them, for example, in cell culture medium containing trypto- lism of polycyclic aromatic hydrocarbons and other environ-
phan, which was recognized as an experimental factor when mental pollutants to which humans are exposed; if so, this
comparing the suitability of commercial cell culture media could play a role in both activating and eliminating topical
with different tryptophan concentrations for Th17 differentia- carcinogenic substances. The balance might be critical [28]. In
tion, which is dependent on AHR [24]. One of these indoles, this context, it is interesting that KC and LC are differentially
1-(1H-indol-3-yl)-9H-pyrido[3,4-b]indole, was originally iso- efficient in xenobiotic metabolism, and the role of the UV-
lated in marine organisms [25, 26]. AHR signaling axis in relation to sensitivity against chemical-
FICZ is similar to TCDD in its lipophilicity, planarity, and induced cancer in this is unexplored [29]. Equally speculative,
shape. Its high-AHR affinity and easy degradability suggest it UV-generated FICZ might also provide an explanation for the
is an endogenous ligand. FICZ is degraded by CYP1A1, thus photosensitivity seen in lupus erythematosus. Conceivably,
regulates its own intracellular steady-state levels. Thus, the generation of FICZ could change the balance of regulatory T
cells toward inflammatory T cells (see below) [30]. Finally,
pigmentation protects the body against DNA damage by UV
Table 2 AHR ligands generated locally in/on skin radiation, and AHR as a sensor for UV light is involved in this
Ligand Source Evidence in skin vital function. In humans, sulfate conjugates of the compound
pathology or health FICZ were identified in 24-h urine [23], suggesting that they
(reference) may be active not only in the skin but in the body as well. It
will be interesting to see, if FICZ is an essential AHR ligand,
FICZ UV plus tryptophan (Sun) Graft rejection [133]
which—similar to Vitamin D—forms in the skin, but is need-
Kynurenine IDO metabolizes tryptophan Vitiligo [134]
(Langerhans cells)
ed “vitamin-like” not only in situ, but also in the whole body.
ICZ Yeast (Malassezia spec) Pityriasis versicolor,
Tryptanthrin, Yeast (Malassezia spec) cancer, [40, 135]
indirubin Kynurenines generated in inflammatory situations
Malassezin Yeast (Malassezia spec). Seborrheic dermatitis,
melanocyte apoptosis
[135, 136]
Catabolic breakdown of tryptophan generated via the so-
called kynurenine pathway leads to several neuroactive and
Semin Immunopathol (2013) 35:677–691 681

immunomodulatory metabolites. More to the point, several production of AHR ligands by Malassezia might contribute to
kynurenines are potent AHR ligands, which have a role in the the risk of skin cancer, such as basal cell carcinomas via its
adaptive differentiation of regulatory T cells [31]. While much effects on cell cycle, DNA repair, reactive oxygen species
is yet unclear, it was recently shown that human dermal (ROS) production, induction of inflammatory cytokines, and
fibroblasts and macrophages fully express the enzymes of by its potential to suppress the immune system [40]. Further
the kynurenine pathway [32]. The immunosuppressive en- work is needed to explore the role of Malassezia-derived
zyme indoleamine 2,3-dioxygenase (IDO) catalyzes the first AHR ligands in skin pathology.
and rate-limiting step in tryptophan catabolism, generating
kynurenine. Recently, we showed that the ido gene is a target
of AHR in Langerhans cells [33], other authors found that AHR involvement in toxic and physiological skin
TCDD induces IDO in DCs derived from bone marrow cells responses
and that such DC promote Treg generation [31, 34]. Interest-
ingly, AHR-deficient dendritic cells do not upregulate IDO In the following, we will discuss the role of AHR for toxic
after stimulation with LPS or IFNγ, and gene expression responses against environmental pollutants, as well as the
profiling of Langerhans cells from AHR-deficient versus wild roles of AHR in healthy skin biology. Not surprisingly, the
type mice showed a virtual shutdown of ido transcription and roles of AHR are as diverse as the functions of the cells in
IDO enzyme activity by AHR deficiency [33, 34]. IDO can be which AHR is expressed in the skin cells. Tables 3 and 4 give
upregulated by proinflammatory cytokines, such as IFNγ or an overview on this spectrum, and indicate where conse-
TNFα, which in turn can be triggered in the skin upon UVB quences for human skin diseases were experimentally/
exposure. epidemiologically addressed or discussed on the background
The physiological consequence of this is currently not of murine data. The therapeutic potential is obvious, although
known, but it was suggested that IDO and tryptophan catab- no clinical trials are published as yet. However, various AHR
olites, especially kynurenine, might be involved in the forma- modulators are on trial for use against breast cancer [41, 42].
tion of regulatory T cells and thus confer tolerance [31, 35, In our institute, studies are under way to treat vitiligo by AHR
36]. Interestingly, one study reported reduced tryptophan manipulation.
levels in vitiligo skin, a common human depigmentation
disease [37]. AHR is vital to the regulation of melanogenesis Chloracne—the most striking human skin response
and melanocyte proliferation and differentiation through mod- against TCDD and other PAHs
ulating the expressions of melanogenesis-related genes.
Thus, one can hypothesize scenarios, whereby sun radia- In humans, “chloracne” is the most striking toxic phenomenon
tion via FICZ, AHR, and IDO sets off the kynurenine path- [43] after TCDD exposure, albeit its intensity does not strictly
way, eventually modulating the local and systemic AHR correlate to exposure dose. Acneiform lesions and cysts were
response. Certainly, this line of research, i.e., the manipulation first described in 1899 and named “chloracne” by Karl
of immune responses by AHR ligands, will attract more Herxheimer, a famous Jewish dermatologist, who later died
attention in the future. in the concentration camp of Theresienstadt when he was
81 years old [44]. Chloracne lesions are caused by acute or
Indole compounds generated by skin-residing yeasts, chronic exposure to dioxins, furans of biphenyls. Albeit great
Malassezia spec improvements have been made to eliminate these substances
from the environment, cases of chloracne still occur to date
The stratum corneum can be colonized by various species of a [45]. The mechanisms underlying chloracne are not well un-
lipophilic yeast, Malassezia , e.g., Malassezia furfur and derstood. Its pathology is characterized by hyperkeratinization
Malassezia globosa. These Malassezia yeasts are present in as well as a metaplastic response of the sebaceous glands.
75∼80 % of healthy subjects, and besides of man, they are Recently, the TCDD-induced skin lesions were recognized as
also present on other warm-blooded animals. Many hamartomas which develop in parallel to a severe atrophy of
Malassezia sp. convert tryptophan into indole compounds sebaceous glands. TCDD accumulates in these hamartomas,
[38], some of which are very potent AHR agonists. The and cyp1a1 is induced. Moreover, TCDD-dependent changes
production of potent AHR ligands by Malassezia yeasts, such in the transcriptome of the lesional tissue point to inhibition of
as indirubin, indolo[3,2-b]carbazole, tryptanthrin and lipid metabolism [46].
malassezin, has been associated with the hyperproliferation Curiously, mice and other laboratory animals do not devel-
in seborrheic dermatitis and the absence of inflammation in op chloracne-like diseases, albeit their keratinocytes are re-
pityriasis versicolor [39]. Underlying mechanisms could be sponsive to TCDD. The reasons for this—as for other species
AHR-mediated immune modulation and effects on cell cycle differences regarding TCDD toxicity—are entirely unknown.
(see below). Finally, as proposed by the group of Gaitanos, the There are no data available on murine sebocytes and AHR. It
682 Semin Immunopathol (2013) 35:677–691

Table 3 Involvement of AHR and its ligands to skin cell and human disease

Skin cell type Evidence in human/mouse Outcome Human skin disease Ref.
discussed

Keratinocytes Human cell culture (SCC-12F, NHEK) Changed TGF-α, TGF-β2, and IL8 production Chloracne, Psoriasis, [137, 138]
after AHR ligand exposure palmoplantar pustulosis
Keratinocytes Human cell culture (NHEK, NIKS), Changed gene expression for keratinocyte Chloracne [93, 139, 140]
mouse (C57BL/J6) differentiation after TCDD exposure
Langerhans Mouse (C57BL/6) Impaired LC maturation and weak contact Contact hypersensitivity [33]
cells hypersensitivity in AHR−/− mice
Melanocytes Human cell culture (NHM, FM55, Upregulation of tyrosinase and MMP gene Skin cancer [55, 141]
A2058, HT-144, Bowes, expression after TCDD exposure
SK-MEL-2)
Melanocytes Mouse (C57BL/6, AhR−/−, AhRΔK5Cre) Impaired melanocyte increase in UVB Vitiligo [142]
exposed AHR−/− mice
Sebocytes Human cell culture (SZ95) Affected sebaceous gland differentiation Chloracne [53]
after TCDD exposure
T cells Mouse (C57BL/6, BALB/c) Affected skin graft survival after AHR ligand Skin graft rejection [133]
exposure
T cells Speculation Influence on inflammatory and autoimmune Psoriasis, atopic dermatis, [131]
diseases by AHR dependent IL-22 production sarcoidosis, SLE
Mast cells Primary mouse MC Enrichment in AhR/IL-6 and AhR/IL-17 (Chronic obstructive [128]
Human histology double-positive MCs within bronchial pulmonary disease)
lamina propria.

should be noted that this research also reveals a role for skin as photocarcinogenesis, i.e., from basal cell carcinomas, squa-
an important xenobiotic metabolizing organ, which produces mous carcinomas, and melanomas. Both increased proliferation
and induces cytochromes P450, epoxide hydroxylases, and of melanocytes and enhanced melanogenesis underly the tan-
other enzymes. Both Langerhans cells and keratinocytes are ning response. The sole producers of melanin are the melano-
effective producers of such enzymes [33, 47], and their effi- cytes, which transfer melanin to keratinocytes via their den-
ciency for xenobiotic metabolism is related to its susceptibility drites. Melanogenesis is a highly complex process, involving
for chemically induced cancer as well as detailed in an elegant approximately 150 genes. Keratinocytes provide α-MSH for
study by Modi et al [29]. melanocytes, which activates the cAMP/PKA pathway, even-
tually resulting in tyrosinase transcription by melanocytes and
AHR: a new kid on the block mediating sun-induced melanin production. Clinical observations after toxic exposures
pigmentation suggested a link between AHR activation and melanogenesis.
In Japan in 1968 and in Taiwan in 1979, accidental mass
Tanning/pigmentation by proliferation of melanocytes and in- poisoning incidents occurred from cooking oil contaminated
creased melanin production is the vital response to protect skin with PCBs, amongst which are strong AHR ligands. Exposed
cells against UV radiation-induced DNA damage leading to persons showed increased skin and gingival pigmentation, and

Table 4 Epidemiological or
clinical links between AHR li- Name of disease Evidence Reference
gands, AHR, and human skin
disease Vitiligo Association of AHR promoter [37, 56]
polymorphism with vitiligo in
a Han Chinese population
Differential expression of IDO
and c-kit in vitiligo lesions
Chloracne TCDD exposure Numerous, [45]
Psoriasis Histology, epidemiological association diMeglio et al., JID132, S15,079
(42nd Ann. Europ. Soc. of Dermatol.
Research Meeting), [143]
SLE UV light exacerbates disease [144]
Eosinophil Fasciitis Toxic oil diseases might trigger via [145]
tryptophan/UV/IDO
Enhanced skin Epidemiological correlation with air Vierkötter et al. submitted
aging pollution
Semin Immunopathol (2013) 35:677–691 683

children of mothers exposed to PCBs were born with dark carcinogenic potential of PAH and related environmental pol-
pigmentation of the head, face, and genitals (“cola-babies”) lutants. PAHs can be found in cigarette smoke, combustion-
[48–51]. Also, TCDD caused skin hyperpigmentation [52]. and traffic-derived particulate matter, and coal tar. Thus, our
Biologically, UV radiation, more precisely UVB (290– skin, as a barrier organ, is frequently exposed to these carcino-
320 nm wavelength), is the most important modulator of skin genic chemicals. Although nontoxic per se, PAHs are oxidized
pigmentation. In line with the observations after chemical by certain CYP enzymes, especially AHR-dependent
(toxic) activation of AHR, our own work could show that CYP1A1, CYP1A2, CYP1B1, and CYP2S1 [3], to enhance
melanocytes functionally express AHR, and the mice needed their water solubility and enable conjugation to hydrophilic
AHR to tan efficiently in standard UVB protocols. Both en- moieties, such as activated sugar, sulfate, or glutathione. In case
hanced cell proliferation and tyrosine-induction were less effi- the capacity of the conjugating enzyme system is exhausted,
cient in AHR knockout mice compared to wild-type mice. PAH metabolites may attack the DNA to form highly muta-
Tanning responses and tyrosinase activity, however, were nor- genic adducts, which may be the initiating step for carcinogen-
mal in keratinocyte-specific conditional AHR knockout mice, esis. In addition, CYP-mediated reactions can cause a higher
indicating that the release of melanogenic keratinocyte factors production of ROS, resulting in oxidative damage of DNA [62]
are unaffected by the UVB-AHR signaling pathway and that and other macromolecules [63]. By now, it is broadly accepted
the diminished tanning response in AHR knockout mice is that cutaneous exposure to PAHs or PAH-containing formula-
confined to the level of melanocytes. It is well known that the tions can give rise to squamous cell carcinomas [64].
c-kit/stem cell factor system is involved in melanocyte homeo- The probably best examined model of PAH relevant for
stasis, and c-kit has functional XREs in its promoter which are human exposure is B[a]P, which is sequentially metabolized
addressed by AHR [53, 54]. Melanogenesis was also inducible by CYP1A1 and microsomal epoxide hydrolase 1 (EPXH1) to
in human cultured melanocytes by TCDD and FICZ [55]. B[a]P-7,8-dihydrodiol-9,10-epoxide, an ultimate carcinogen
Thus, the environmental sensor AHR links solar UVB radiation [65]. CYP1A1 expression is abolished in AHR-deficient
to skin pigmentation. This finding is relevant for clinical appli- mice, and these animals are completely protected against
cations, as it opens up the possibility of manipulating melano- B[a]P carcinogenicity [66].
cyte proliferation (rather than mere chemical stimulation of The metabolic activation of DMBA, another model PAH,
tyrosinase) as a therapeutic approach in diseases such as vitili- is mediated by CYP1A1, CYP1B1, and EPXH1 [67]. In a
go, a depigmentation disorder, or cosmetic problems such as classical two-stage carcinogenesis study using 12-O-
solar-induced lentigines in aging skin. Of note, a promoter tetradecanoylphorbol-13-acetate as promoter, DMBA-exposed
polymorphism of AHR in humans was linked to increased risk mice developed cutaneous squamous cell carcinomas [29, 68].
for vitiligo as studied in a Han Chinese population [56], and The vast majority of these tumors harbored the activating codon
low c-kit levels are detected in vitiligo lesions and their periph- 61 mutation in the Hras proto-oncogene [69]. Interestingly, the
eral areas [57]. carcinogenic potential of DMBA strongly depends on which
CYP1 isoform is predominantly expressed in the exposed cell
The AHR is a central mediator of chemical skin population or tissue [67]. In contrast to CYP1A1-mediated
carcinogenesis DMBA metabolism (which favors detoxification), CYP1B1-
mediated metabolism of DMBA results in an enhanced forma-
The oncogenic potential of the AHR was elegantly demonstrat- tion of DMBA-trans-3,4-diol [70–72]. This metabolite is sub-
ed using a transgenic mouse line that expresses a constitutively sequently transformed to the highly mutagenic DMBA-3,4-
active form of AHR. In contrast to their AHR-proficient litter- diol-1,2-epoxide. In this context, a recent study revealed that
mates, these transgenic mice developed severe tumors in the in keratinocytes, CYP1A1 is clearly higher expressed than
glandular part of the stomach in absence of any additional CYP1B1, whereas the latter is the dominating CYP enzyme
initiators or promoters [58]. Furthermore, a single injection in Langerhans cells. Experimental depletion of epidermal
of N -nitrosodiethylamine was sufficient to induce Langerhans cells protected mice against DMBA-induced skin
hepatocarcinogenesis in these transgenic animals [59], revealing carcinogenesis, providing evidence that the generation of
the tumor promoting properties of an overactivated AHR sys- DMBA-3,4-diol-1,2-epoxide is mediated exclusively by epi-
tem. These are probably due to AHR-dependent activation of dermal Langerhans cells [29]. The basal and inducible expres-
mitogenic and proinflammatory cytokines and growth factors, sion of CYP1B1 is not only regulated by the AHR, but also by
such as COX-2, IL-8, IL-18, and amphiregulin [60, 61]. Mean- other factors such as estrogen receptor-α [73] or effectors of
while, numerous other publications also contributed to the activated protein kinase A [74]. Accordingly, the rate of
awareness that AHR triggers signaling processes relevant for CYP1B1 expression is still high enough in AHR KO mice to
the development and progression of different types of cancer. toxify DMBA. Consequently, AHR KO mice were shown to
In the context of skin carcinogenesis, the AHR and down- develop squamous cell carcinomas in comparable amounts to
stream CYP enzymes were proven to be essential to unleash the AHR wild-type mice [67].
684 Semin Immunopathol (2013) 35:677–691

Although reports exist showing that CYP1-deficient mice receptors (TLRs). They are pivotal in mobilizing leukocytes
exhibit higher amounts of B[a]P-DNA adducts than the respec- from blood and orchestrate the participation of dermal cells in
tive wild-type animals [75], a timely limited inhibition of the an immune response. AHR was identified as a crucial medi-
AHR may probably result in a reduced metabolic activation of ator of the UVB stress response in human keratinocytes,
environmental pollutants, and thus inhibit chemical carcino- which triggers activation of the EGFR and downstream
genesis. Regarding the usage of AHR antagonists as chemo- MAPK cascades upon UVB exposure. Activation of MAPK
preventive, compounds that attenuate ligand-activated AHR is relevant for UVB-induced skin inflammation and
signaling and simultaneously stimulate the Nrf2 system are photocarcinogenesis [85]. Again, FICZ is the relevant ligand.
probably promising candidates to prevent severe health effects It was an exciting finding that AHR might not only trigger the
provoked by environmental stressors [76]. As the master reg- classical nuclear events of DNA transcription. Thus, dissolu-
ulator of the antioxidant response, Nrf2 controls the expression tion of the cytosolic AHR complex releases c-src, which in
of numerous phase II conjugating enzymes, which are capable turn binds to EGFR and initiates MAPK signaling, resulting in
of neutralizing ROS and reactive phase I metabolites [77]. the upregulation of target genes, e.g., COX-2 [8]. Interesting-
Indeed, it was shown that co-exposure of mice to sulforaphane, ly, EGFR activation inhibits many processes in keratinocyte
a dietary isothiocyanate, protected the mice against DMBA- differentiation, and was shown in organotypic cultures of
induced skin tumorigenesis via activation of Nrf2 signaling human skin to impair epidermal integrity. Many EGFR-
[78], underscoring the importance of the capacity of the phase targeted genes are related to skin diseases [86]. Sutter and
II system in restraining chemical skin carcinogenesis. coworkers recently reported that EGF represses the dioxin-
Besides its involvement in chemical carcinogenesis, the fact mediated induction of CYP1A1 in cultured normal human
that the AHR is activated in response to UVB exposure implies keratinocytes by inhibiting the recruitment of the transcrip-
its possible contribution to photocarcinogenesis. The UVB- tional coactivator protein p300 to the CYP1A1 gene. TCDD
stimulated activation of AHR in keratinocytes results in an accelerates keratinocyte differentiation and the formation of
increased expression of CYP1A1, CYP1B1, and COX-2 [8, the cornified envelope [87], and blocking or manipulating
27], whose enzyme-products may trigger photocarcinogenesis EGFR signaling was suggested as a therapeutic option in
[79]. Indeed, topical application of some plant polyphenols dioxin-like skin pathologies.
that are known to antagonize AHR signaling, for instance Although a direct study on AHR-mediated EGFR trigger-
resveratrol or epigallocatechin-3-gallate [80, 81], were shown ing in skin diseases has not been done so far, the data
to protect mice against UVB-induced skin carcinogenesis [82, and observations fit well with the results from a mouse
83]. Moreover, recent results from our laboratory disclose an model which express a constitutively activate AHR in its
antiapoptotic function of the AHR in UVB-exposed human keratinocytes. This mouse line develops severe skin lesions
and murine keratinocytes. Accordingly, a chemical inhibition with itching, skin inflammation, and immunological imbal-
of AHR led to an enhanced removal of cells harboring irrep- ance. Together, this resembled typical atopic dermatitis, a
arable DNA damage (T.H.-S., unpublished observations). chronic inflammatory disorder characterized by a defect in
Since apoptosis is regarded as one of the most important epidermal barrier function [61]. Also, keratinocyte-specific
mechanisms restraining carcinogenesis [84], these results ARNT-deficient mice die shortly after birth due to severe
again point to a critical role of the AHR during skin cancer dehydration, caused by a defect in normal lipid production
development, which warrants further investigation. by keratinocytes [88].
What about AHR and cytokines in keratinocytes?
Keratinocytes express an impressive arsenal of cytokines
AHR and the skin immune system [89]. They produce proinflammatory ones such as IL1β
or TNFα, as well as those controlling growth (IL-6, stem
Immunity against pathogens on the skin while not attacking cell factor), and immunosuppressive cytokines (TGF-β),
the harmless commensals on the skin, immunosurveillance chemokines (IL-8), and more [90], many of which have XREs
against cancer cells and wound healing are important func- in their promoters [4, 91]. Direct evidence shows that IL1β
tions of the skin. There are both resident antigen-presenting [92] and IL-8 are targets of AHR. IL-8 is induced in human
cells and T cells in the skin, and during an immune response, keratinocytes by B[a]P—a chemical found also in cigarette
more cells immigrate, guided, e.g., by chemokine gradients. smoke—and IL-8 is related to inflammatory skin diseases
AHR is relevant in all of these tasks. [93]. In wound healing, levels of TGF-β are decisive for
epithelization and migration of keratinocytes to close wounds.
AHR and the keratinocyte barrier function Delayed wound healing causes chronic skin lesions such as
those found in diabetes. Fernandez-Salguero and his group
Keratinocytes are the first line of defense in the skin immune showed in AHR-deficient mice that an increased TGF-β
system. They secrete many cytokines and possess toll-like production by fibroblasts led to faster re-epithelization and a
Semin Immunopathol (2013) 35:677–691 685

higher collagen content. Possibly, down-modulation of AHR through its capacity to degrade the essential amino acid tryp-
could be used to improve wound healing [94]. tophan into kynurenine and other downstream metabolites,
which suppress effector T cell function and favor the differen-
AHR and dendritic cells of the skin tiation of regulatory T cells. DC and certain tumors can express
the ido gene. IDO-mediated tolerogenicity and acquired im-
LC make up 1–3 % of the epidermal cells. As DC, they mune privilege has been identified as a potential target for
acquire, process, and subsequently present both foreign and therapeutical strategies in inflammatory and autoimmune dis-
self-antigens to T lymphocytes in lymphoid organs. Several eases, such as collagen arthritis or diabetes [53, 56]. Unexpect-
years ago, in a shift of paradigm, evidence emerged that LC— edly, IDO enzyme activity was not inducible by LPS or IFN-γ
beyond initiating contact hypersensitivity [95]—have regula- in bone marrow-derived DC from AHR-deficient mice [33,
tory and tolerogenic functions [96]. Furthermore, LC was 34]. As AHR is known to interact with NFκB [104], it is likely
shown to be unable to elicit T cell responses to certain viral that ido induction by AHR does not use the “classical” AHR/
antigens [97, 98]. In 1989, Puhvel et al. [99] investigated ARNT pathway described above, but ties into NFκB signaling
density and morphology of LC in HRS/J mice, a murine [7, 105]. Data for dermal DC are not available. What could be
model for skin effects of TCDD. They reported that LC from the consequence of IDO as a target of AHR in LC/DC?
TCDD-treated hairless mice were smaller and had fewer den- Kynurenines produced by DC can induce Treg directly [31],
dritic protrusions than controls. We showed that LC and this might explain how DC can promote Treg formation in
Langerhans cells express AHR (and also AHRR) at high an AHR-dependent manner. Currently, this was shown only
levels [33]. AHR-deficient LC was impaired in maturation; in vitro, but conceivably, it could be a previously unrecognized
they remained smaller and less granular, and did not mechanism on how the skin calms overshooting inflammatory
upregulate expression of co-stimulatory molecules CD40, reactions. At the same time, the ligand FICZ promotes gener-
CD80, and CD24 during in vitro maturation. Likewise, their ation of Th17 from naive T cells [24, 106, 107]. Interestingly,
phagocytic capacity did not decrease (another sign of imma- in human atopic skin or psoriatic lesional skin, the levels of ido
turity). GMCSF, needed for LC maturation, was secreted in are higher than in uninvolved skin [108]. It is currently difficult
significantly lower amounts by AHR-deficient epidermal to solve these seemingly contradictory findings, and more
cells, largely due to invariant γδ T cells [100]. The data research is needed before designing therapeutic strategies for
suggests that the AHR is involved in LC maturation, both skin diseases [109].
cell-autonomously and through by-stander cells. TCDD injec-
tion of mice did not lead to upregulation of CYP1A1 in LC; AHR and IL17, IL22, and IL23
this might be related to the high constitutive levels of AHRR
and might be part of a risk strategy against protein-reactive As was first shown by two seminal papers in 2008 [106, 107]
generation of chemical skin allergens [101, 102]. and soon studied in increasing details, AHR is a necessary
Contact hypersensitivity is impaired in AHR deficient factor for the production of IL-22 by Th17 T cells, and for the
mice, which is in agreement with the impaired maturation of differentiation balance of Th17 and/or inducible Foxp3+ Treg
AHR-deficient LC. We have observed that AHR−/−LC retain and the generation of Tr1 cells [24, 105, 107, 110, 111]. Th17
their capacity to migrate into LN after hapten challenge (C.E., cells are a new effector subset of T cells, and pivotal for
unpublished observation). Contact hypersensitivity is compa- fighting bacteria. They also contribute to the exacerbation of
rable between WT mice and mice with conditional ablation of autoimmune diseases; thus, it is essential that they are tightly
AHR in KC only, suggesting that the impairment is caused by controlled. AHR activity promotes their expansion and is
an intrinsic LC effect (C.E., unpublished data). Blocking of obligatory for IL-22 production. By now, the picture is at the
the AHR signal in LC may help to relieve allergic skin symp- same time more detailed and infinitely more complex; in
toms because many xenobiotics, including those metabolized particular, it emerged that AHR also promotes IL21 and
via the AHR pathway, can exacerbate allergic reactions [102]. IL23 production and has, dependent on the ligand, also inhib-
Transgenic mice with a constitutively active AHR in KC itory effects on Th17 [31]. The skin has several subsets of IL-
develop skin lesions accompanied by inflammation and im- 17 producing cells (see also below), which may not be sur-
munological imbalances resembling typical atopic dermatitis prising considering for a site of constant bacterial exposure.
[61], and TCDD exacerbates atopic dermatitis [103]. The Also in γδ T cells, IL-22 is controlled by AHR activity [13]. In
underlying mechanism is not clear, albeit the triggering events humans, LC induces a special subset of T cells, Th22, which
might be a break of the skin barrier, followed by inflammation. produce IL-22 but not IL-17 [112]. Again, IL-22 is under the
A second highly interesting angle is the role of AHR for control of AHR [113]. IL-22 participates in many chronic
IDO expression, both by LC and bone marrow-derived DC. inflammatory conditions, including in the skin. It promotes
Both constitutive and inducible expression of IDO requires KC hyperplasia and epidermal remodeling in 3D cultures, and
presence of AHR in the cells. IDO regulates immune responses has been suggested as a target in the treatment of psoriasis
686 Semin Immunopathol (2013) 35:677–691

[114]. Many questions remain, both regarding the AHR- are a major source of IL17 in skin [123]. Also, Th17 cells are
dependent regulation of these cytokines in skin homeostasis found in psoriasis and atopic dermatitis, and are pivotal for
and during skin pathologic disease conditions, as well as the defense against bacterial and fungal infections [13].
associated therapeutic options. Regarding the role of AHR in Considering the role of γδ T cells in skin immuno-
changing the balance of regulatory T cells versus Th17 cells, surveillance, and their responsiveness to AHR signaling, it
AHR has been suggested as a target to combat autoimmune will be exciting to follow further how environmental cues
disorders [111, 115]. Eventually, it will be important to study such as UV light (generating FICZ) or TLR activation by
whether and how skin-resident T cells are sensitive to ligand- commensal or pathogenic bacteria (possibly connecting
mediated AHR activation in situ, and whether their plasticity AHR to NFκB signaling) triggers and shapes skin immunity.
is affected by AHR, or whether the fate of T cells which
immigrate into skin upon inflammation can be manipulated
by AHR.
Conclusions and outlook
AHR and skin gamma–delta cells
Research on AHR as a sensor linking the environment to
The murine epidermis harbors a special subset of TCRγδ + T physiological functions has moved with impressive speed
cells with a restricted repertoire of Vγ3/Jγl-Cγ1 and Vδ1/Jδ2/ over the last years, far beyond the decade-long studies in
Jδ2-Cδ (according to the nomenclature by Garmann, Cell toxicology of the AHR. In particular, immunological and
1986). These skin TCRγδ + T cells are also called dendritic oncological questions are studied. It was soon recognized that
epidermal T cells (DETC) because of their morphology. Mice there is a strong ligand-dependent and cell-type dependent
deficient in the tcrd gene, and thus without any TCRγδ + complexity in outcome of AHR activation, in addition to
(neither in the periphery nor in the skin), have increased dose-effects, the bifurcated quality of signaling via c-src in
sensitivity to carcinogen-induced skin carcinogenesis and the cytoplasm and AHR/ARNT, and finally its potential to tie
wound repair [116]. Interestingly, most γδ T cells are an into other signaling pathways such as NFκB. This paper, and
important source of IL17 and IL22, and—similar to Th17 the other ones in this special issue of Seminars in Immunopa-
cells—AHR is required for IL22 production [117]. As they thology, gives an idea of the impressive range of AHR effects
express TLRs, they are uniquely suited for quickly fighting and its underlying cellular events. Unfortunately, AHR has not
bacterial infections, before the adaptive immune response been crystallized until now, hampering structure-activity stud-
kicks in. High AHR expression has been shown in murine ies of AHR and its ligands. This certainly is a huge gap in
DETC, gut intraepithelial lymphocytes, and in a subset of AHR research.
peripheral IL-17 producing TCRγδ + T cells [100, 117, Nonetheless, AHR is uniquely suited for therapeutic ap-
118]. We found that DETC can seed the skin after birth, but proaches or chemopreventive strategies. This type of research
do not proliferate in situ and are quickly lost in AHR-deficient is most advanced regarding cancer such as breast cancer and
mice. In contrast to tcrd −/− mice (which are γδ T cell deficient brain tumors. The group of Stephen Safe has tested pharma-
in the periphery as well), DETC are not replaced by αβ T ceuticals already in clinical use, which have AHR agonist
cells, making AHR-deficient mice a unique model to study capacity (e.g., leflunomide, tranilast, 4-hydroxytamoxifen,
DETC function [100]. The molecular mechanism for this loss omeprazole, and others). They and others reported the effects
is not altogether clear, but evidence points to the reduced on breast cancer cells suggesting their clinical potential for
expression of c-kit, which is a direct target of AHR, and treatment of breast cancer via AHR-ER inhibitory cross-talk
needed for γδ T cell homeostasis. DETC are a major source [41, 124] and have labeled such molecules selective AHR
of GMCSF and other cytokines in the skin, their lack affects modulators. Interestingly, tranilast is also used in treatment
LC and KC as well [116]. of atopic dermatitis. Tryptophan degradation/kynurenine gen-
Human skin has more TCRαβ + T cells and fewer γδ T eration by brain tumor cells appears to be a strategy to escape
cells than murine skin. Also in human skin, γδ T cells secrete the immune system in an AHR-dependent manner, and this
cytokines, are important immunosurveillance [119, 120], and novel mechanism might have broader implications in the
presumably participate in the control of epithelial integrity relationship of inflammation and cancer in general [125]. In
[121]. The human-resident T cells are mainly memory-type particular, FICZ was considered a novel target [126], and plant
CD8+ αβTCR + T cells in the epidermis and cutaneous polyphenols (e.g., green tee polyphenols, flavonoids, and
lymphocyte antigen expressing CD4+ and CD8+ T cells in many more) have been studied for their use in skin cancer,
the dermis [12, 13]. Conventional T cells of the Th1, Th2, and as recently reviewed by Korkina and coworkers [127]. Re-
Th17 type migrate into the skin during various inflammatory garding therapy, suitable AHR ligands can be integrated into
diseases. Recently, it was found that invariant Vγ9 T cells creams without the solvent or logistic problems associated
migrate from blood into psoriatic lesions [122], and γδ T cells with large protein biologics.
Semin Immunopathol (2013) 35:677–691 687

In conclusion, AHR is an important player in skin physi- competitive polymerase chain reaction to measure Ah-receptor
mRNA expression. Arch Biochem Biophys 315(2):279–284
ology, and although much is still unclear, AHR emerges as a
16. Esser C (2012) The physiological role of AHR in the mouse
target for prevention and therapy of skin diseases. It will be immune system. In: Pojhanvirta R (ed) The AH receptor in biology
exciting to see more and more data, both from basic research and toxicology. Ref Type: Serial (Book,Monograph). Wiley&Sons,
as from clinical studies in the near future. Hoboken, pp 499–510
17. Denison MS, Nagy SR (2003) Activation of the aryl hydrocarbon
receptor by structurally diverse exogenous and endogenous
Acknowledgments Research of C.E. is supported by grants from the chemicals. Annu Rev Pharmacol Toxicol 43:309–334
Deutsche Forschungsgemeinschaft (DFG ES103/5-1 and 103/6-1). H.W. 18. Nguyen LP, Bradfield CA (2008) The search for endogenous acti-
is supported by DFG WE2625/2-1 and the Leibnizgemeinschaft vators of the aryl hydrocarbon receptor. Chem Res Toxicol
(SenatsausschussWettbewerb 2012). J.K. is supported by DFG FG871 21(1):102–116
and the ‘Bundesministerium für Bildung und Forschung Verbundprojekt 19. Denison MS, Soshilov AA, He G, DeGroot DE, Zhao B (2011)
UV’. Exactly the same but different: promiscuity and diversity in the
molecular mechanisms of action of the aryl hydrocarbon (dioxin)
receptor. Toxicol Sci 124(1):1–22
20. Rannug U, Rannug A, Sjoberg U, Li H, Westerholm R, Bergman J
References (1995) Structure elucidation of two tryptophan-derived, high affin-
ity Ah receptor ligands. Chem Biol 2(12):841–845
21. Rannug A, Rannug U, Rosenkranz HS, Winqvist L, Westerholm R,
1. Schmidt JV, Bradfield CA (1996) Ah receptor signaling pathways. Agurell E et al (1987) Certain photooxidized derivatives of trypto-
Annu Rev Cell Dev Biol 12:55–89 phan bind with very high affinity to the Ah receptor and are likely to
2. Dolwick KM, Schmidt JV, Carver LA, Swanson HI, Bradfield CA be endogenous signal substances. J Biol Chem 262(32):15422–
(1993) Cloning and expression of a human Ah receptor cDNA. Mol 15427
Pharmacol 44(5):911–917 22. Diani-Moore S, Labitzke E, Brown R, Garvin A, Wong L, Rifkind
3. Abel J, Haarmann-Stemmann T (2010) An introduction to the AB (2006) Sunlight generates multiple tryptophan photoproducts
molecular basics of aryl hydrocarbon receptor biology. Biol Chem eliciting high-efficacy CYP1A induction in chick hepatocytes and
391(11):1235–1248 in vivo. Toxicol Sci 90(1):96–110
4. Sun YV, Boverhof DR, Burgoon LD, Fielden MR, Zacharewski TR 23. Wincent E, Amini N, Luecke S, Glatt H, Bergman J, Crescenzi C
(2004) Comparative analysis of dioxin response elements in human, et al (2009) The suggested physiologic aryl hydrocarbon receptor
mouse and rat genomic sequences. Nucleic Acids Res 32(15):4512– activator and cytochrome P4501 substrate 6-formylindolo[3,2-b]
4523 carbazole is present in humans. J Biol Chem 284(5):2690–2696
5. Frericks M, Meissner M, Esser C (2007) Microarray analysis of the 24. Veldhoen M, Hirota K, Christensen J, O’Garra A, Stockinger B
AHR system: tissue-specific flexibility in signal and target genes. (2009) Natural agonists for aryl hydrocarbon receptor in culture
Toxicol Appl Pharmacol 220(3):320–332 medium are essential for optimal differentiation of Th17 T cells. J
6. Mitchell KA, Elferink CJ (2009) Timing is everything: conse- Exp Med 206(1):43–49
quences of transient and sustained AhR activity. Biochem 25. Diani-Moore S, Ma Y, Labitzke E, Tao H, David WJ, Anderson J
Pharmacol 77(6):947–956 et al (2011) Discovery and biological characterization of 1-(1H-
7. Wu D, Li W, Lok P, Matsumura F, dam Vogel CF (2011) AhR indol-3-yl)-9H-pyrido[3,4-b]indole as an aryl hydrocarbon receptor
deficiency impairs expression of LPS-induced inflammatory genes activator generated by photoactivation of tryptophan by sunlight.
in mice. Biochem Biophys Res Commun 410(2):358–363 Chem Biol Interact 193(2):119–128
8. Fritsche E, Schafer C, Calles C, Bernsmann T, Bernshausen T, 26. Hahn ME (2002) Aryl hydrocarbon receptors: diversity and evolu-
Wurm M et al (2007) Lightening up the UV response by identifi- tion. Chem Biol Interact 141(1–2):131–160
cation of the arylhydrocarbon receptor as a cytoplasmatic target for 27. Katiyar SK, Matsui MS, Mukhtar H (2000) Ultraviolet-B exposure
ultraviolet B radiation. Proc Natl Acad Sci USA 104(21):8851– of human skin induces cytochromes P450 1A1 and 1B1. J Invest
8856 Dermatol 114(2):328–333
9. Mimura J, Ema M, Sogawa K, Fujii-Kuriyama Y (1999) Identifi- 28. Ma Q, Lu AY (2007) CYP1A induction and human risk assessment:
cation of a novel mechanism of regulation of Ah (dioxin) receptor an evolving tale of in vitro and in vivo studies. Drug Metab Dispos
function. Genes Dev 13(1):20–25 35(7):1009–1016
10. Tigges J, Weighardt H, Wolff S, Gotz C, Forster I, Kohne Z, et al. 29. Modi BG, Neustadter J, Binda E, Lewis J, Filler RB, Roberts SJ et al
(2012) Aryl Hydrocarbon Receptor Repressor (AhRR) function (2012) Langerhans cells facilitate epithelial DNA damage and squa-
revisited: repression of CYP1 activity in human skin fibroblasts is mous cell carcinoma. Science 335(6064):104–108
not related to AhRR expression. J Invest Dermatol 30. Martin R, O’Shea J, Birnbaum LS, Luebke R (2008) Community
11. Romani N, Clausen BE, Stoitzner P (2010) Langerhans cells and corner. Striking the balance in multiple sclerosis. Nat Med
more: langerin-expressing dendritic cell subsets in the skin. 14(5):491
Immunol Rev 234(1):120–141 31. Mezrich JD, Fechner JH, Zhang X, Johnson BP, Burlingham WJ,
12. Clark RA, Chong B, Mirchandani N, Brinster NK, Yamanaka K, Bradfield CA (2010) An interaction between kynurenine and the
Dowgiert RK et al (2006) The vast majority of CLA + T cells are aryl hydrocarbon receptor can generate regulatory T cells. J
resident in normal skin. J Immunol 176(7):4431–4439 Immunol 185(6):3190–3198
13. Nestle FO, Di MP, Qin JZ, Nickoloff BJ (2009) Skin immune 32. Asp L, Johansson AS, Mann A, Owe-Larsson B, Urbanska EM,
sentinels in health and disease. Nat Rev Immunol 9(10):679–691 Kocki T et al (2011) Effects of proinflammatory cytokines on
14. Carlstedt-Duke JM (1979) Tissue distribution of the receptor for expression of kynurenine pathway enzymes in human dermal fibro-
2,3,7,8-tetrachlorodibenzo-p-dioxin in the rat. Cancer Res blasts. J Inflamm (Lond) 8:25
39(8):3172–3176 33. Jux B, Kadow S, Esser C (2009) Langerhans cell maturation and
15. Li W, Donat S, Dohr O, Unfried K, Abel J (1994) Ah receptor in contact hypersensitivity are impaired in aryl hydrocarbon receptor-
different tissues of C57BL/6 J and DBA/2 J mice: use of null mice. J Immunol 182(11):6709–6717
688 Semin Immunopathol (2013) 35:677–691

34. Nguyen NT, Kimura A, Nakahama T, Chinen I, Masuda K, Nohara 51. Hashiguchi I, Yoshimine Y, Maeda H, Gotou Y, Ishikawa M, Fujii S
K et al (2010) Aryl hydrocarbon receptor negatively regulates et al (2007) Epidemiologic examination on the prevalence of the
dendritic cell immunogenicity via a kynurenine-dependent mecha- periodontal diseases and oral pigmentation in Yusho patients in
nism. Proc Natl Acad Sci U S A 107(46):19961–19966 2006. Fukuoka Igaku Zasshi 98(5):170–175
35. Mellor AL, Munn DH (2004) IDO expression by dendritic cells: 52. Dunagin WG (1984) Cutaneous signs of systemic toxicity due to
tolerance and tryptophan catabolism. Nat Rev Immunol 4(10):762– dioxins and related chemicals. J Am Acad Dermatol 10(4):688–700
774 53. Jux B, Kadow S, Luecke S, Rannug A, Krutmann J, Esser C (2011)
36. Frumento G, Rotondo R, Tonetti M, Damonte G, Benatti U, Ferrara The aryl hydrocarbon receptor mediates UVB radiation-induced
GB (2002) Tryptophan-derived catabolites are responsible for inhi- skin tanning. J Invest Dermatol 131(1):203–210
bition of T and natural killer cell proliferation induced by 54. Kiss EA, Vonarbourg C, Kopfmann S, Hobeika E, Finke D, Esser C
indoleamine 2,3-dioxygenase. J Exp Med 196(4):459–468 et al (2011) Natural aryl hydrocarbon receptor ligands control
37. Schallreuter KU, Salem MA, Gibbons NC, Maitland DJ, Marsch E, organogenesis of intestinal lymphoid follicles. Science. doi:10.
Elwary SM et al (2012) Blunted epidermal L -tryptophan metabo- 1126/science.1214914
lism in vitiligo affects immune response and ROS scavenging by 55. Luecke S, Backlund M, Jux B, Esser C, Krutmann J, Rannug A
Fenton chemistry, part 2: epidermal H2O2/ONOO(−)-mediated (2010) The aryl hydrocarbon receptor (AHR), a novel regulator of
stress in vitiligo hampers indoleamine 2,3-dioxygenase and aryl human melanogenesis. Pigment Cell Melanoma Res 23(6):828–833
hydrocarbon receptor-mediated immune response signaling. 56. Wang XW, Li K, Guo S, Qiang HN, Liu L, Song P et al (2012) The
FASEB J 26(6):2471–2485 association of functional polymorphisms in the aryl hydrocarbon
38. Wille G, Mayser P, Thoma W, Monsees T, Baumgart A, Schmitz HJ receptor (AHR) gene with the risk of vitiligo in Han Chinese
et al (2001) Malassezin–A novel agonist of the arylhydrocarbon populations. Br J Dermatol 166(5):1081–1087
receptor from the yeast Malassezia furfur. Bioorg Med Chem 57. Kitamura R, Tsukamoto K, Harada K, Shimizu A, Shimada S,
9(4):955–960 Kobayashi T et al (2004) Mechanisms underlying the dysfunction
39. Vlachos C, Schulte BM, Magiatis P, Adema GJ, Gaitanis G (2012) of melanocytes in vitiligo epidermis: role of SCF/KIT protein
Malassezia-derived indoles activate the aryl hydrocarbon receptor interactions and the downstream effector, MITF-M. J Pathol
and inhibit toll-like receptor-induced maturation in monocyte- 202(4):463–475
derived dendritic cells. Br J Dermatol 167(3):496–505 58. Andersson P, McGuire J, Rubio C, Gradin K, Whitelaw ML,
40. Gaitanis G, Velegraki A, Magiatis P, Pappas P, Bassukas ID (2011) Pettersson S et al (2002) A constitutively active dioxin/aryl hydro-
Could Malassezia yeasts be implicated in skin carcinogenesis carbon receptor induces stomach tumors. Proc Natl Acad Sci U S A
through the production of aryl-hydrocarbon receptor ligands? Med 99(15):9990–9995
Hypotheses 77(1):47–51 59. Moennikes O, Loeppen S, Buchmann A, Andersson P, Ittrich C,
41. Jin UH, Lee SO, Safe S (2012) Aryl hydrocarbon receptor (AHR)- Poellinger L et al (2004) A constitutively active dioxin/aryl hydro-
active pharmaceuticals are selective AHR modulators in MDA-MB- carbon receptor promotes hepatocarcinogenesis in mice. Cancer Res
468 and BT474 breast cancer cells. J Pharmacol Exp Ther 64(14):4707–4710
343(2):333–341 60. Haarmann-Stemmann T, Bothe H, Abel J (2009) Growth factors,
42. Tiong CT, Chen C, Zhang SJ, Li J, Soshilov A, Denison MS et al cytokines, and their receptors as downstream targets of aryl hydro-
(2012) A novel prenylflavone restricts breast cancer cell growth carbon receptor (AhR) signaling pathways. Biochem Pharmacol
through AhR-mediated destabilization of ERalpha protein. Carci- 77(4):508–520
nogenesis 33(5):1089–1097 61. Tauchi M, Hida A, Negishi T, Katsuoka F, Noda S, Mimura J et al
43. Caputo R, Monti M, Ermacora E, Carminati G, Gelmetti C, Gianotti (2005) Constitutive expression of aryl hydrocarbon receptor in
R et al (1988) Cutaneous manifestations of tetrachlorodibenzo-p- keratinocytes causes inflammatory skin lesions. Mol Cell Biol
dioxin in children and adolescents. Follow-up 10 years after the 25(21):9360–9368
Seveso, Italy, accident. J Am Acad Dermatol 19(5 Pt 1):812–819 62. Xue W, Warshawsky D (2005) Metabolic activation of polycyclic
44. Scholz A (2012) Karl Herxheimer - Geheimrat mit dem gelben and heterocyclic aromatic hydrocarbons and DNA damage: a re-
Stern. J Dtsch Dermatol Ges 10(12):925–927 view. Toxicol Appl Pharmacol 206(1):73–93
45. Passarini B, Infusino SD, Kasapi E (2010) Chloracne: still cause for 63. Godschalk R, Curfs D, Bartsch H, van Schooten FJ, Nair J (2003)
concern. Dermatology 221(1):63–70 Benzo[a]pyrene enhances lipid peroxidation induced DNA damage
46. Saurat JH, Kaya G, Saxer-Sekulic N, Pardo B, Becker M, in aorta of apolipoprotein E knockout mice. Free Radic Res
Fontao L et al (2012) The cutaneous lesions of dioxin exposure: 37(12):1299–1305
lessons from the poisoning of Victor Yushchenko. Toxicol Sci 64. Diepgen TL, Mahler V (2002) The epidemiology of skin cancer. Br
125(1):310–317 J Dermatol 146(Suppl 61):1–6
47. Gotz C, Pfeiffer R, Tigges J, Blatz V, Jackh C, Freytag EM et al 65. Gelboin HV (1980) Benzo[alpha]pyrene metabolism, activation,
(2012) Xenobiotic metabolism capacities of human skin in compar- and carcinogenesis: role and regulation of mixed-function oxidases
ison with a 3D epidermis model and keratinocyte-based cell culture and related enzymes. Physiol Rev 60(4):1107–1166
as in vitro alternatives for chemical testing: activating enzymes 66. Shimizu Y, Nakatsuru Y, Ichinose M, Takahashi Y, Kume H,
(Phase I). Exp Dermatol 21(5):358–363 Mimura J et al (2000) Benzo[a]pyrene carcinogenicity is lost in
48. Kikuchi M (1984) Autopsy of patients with yusho. Prog Clin Biol mice lacking the aryl hydrocarbon receptor. Proc Natl Acad Sci U S
Res 137:19–30 A 97(2):779–782
49. Kanagawa Y, Matsumoto S, Koike S, Tajima B, Fukiwake N, 67. Ide F, Suka N, Kitada M, Sakashita H, Kusama K, Ishikawa T
Shibata S et al (2008) Association of clinical findings in (2004) Skin and salivary gland carcinogenicity of 7,12-
Yusho patients with serum concentrations of polychlorinated dimethylbenz[a]anthracene is equivalent in the presence or absence
biphenyls, polychlorinated quarterphenyls, and 2,3,4,7,8- of aryl hydrocarbon receptor. Cancer Lett 214(1):35–41
pentachlorodibenzofuran more than 30 years after the poisoning 68. Hennings H, Devor D, Wenk ML, Slaga TJ, Former B, Colburn NH
event. Environ Health 7:47 et al (1981) Comparison of two-stage epidermal carcinogenesis
50. Masuda Y (1985) Health status of Japanese and Taiwanese after initiated by 7,12-dimethylbenz(a)anthracene or N-methyl-N’-nitro-
exposure to contaminated rice oil. Environ Health Perspect 60:321– N-nitrosoguanidine in newborn and adult SENCAR and BALB/c
325 mice. Cancer Res 41(3):773–779
Semin Immunopathol (2013) 35:677–691 689

69. Balmain A, Ramsden M, Bowden GT, Smith J (1984) Activation of receptor-mediated transcription and cell differentiation in human
the mouse cellular Harvey-ras gene in chemically induced benign epidermal keratinocytes. Proc Natl Acad Sci U S A 106(11):
skin papillomas. Nature 307(5952):658–660 4266–4271
70. Buters J, Quintanilla-Martinez L, Schober W, Soballa VJ, 88. Takagi S, Tojo H, Tomita S, Sano S, Itami S, Hara M et al (2003)
Hintermair J, Wolff T et al (2003) CYP1B1 determines susceptibil- Alteration of the 4-sphingenine scaffolds of ceramides in
ity to low doses of 7,12-dimethylbenz[a]anthracene-induced ovar- keratinocyte-specific Arnt-deficient mice affects skin barrier func-
ian cancers in mice: correlation of CYP1B1-mediated DNA adducts tion. J Clin Invest 112(9):1372–1382
with carcinogenicity. Carcinogenesis 24(2):327–334 89. Matsue H, Cruz PD Jr, Bergstresser PR, Takashima A (1992)
71. Savas U, Carstens CP, Jefcoate CR (1997) Biological oxidations Cytokine expression by epidermal cell subpopulations. J Invest
and P450 reactions. Recombinant mouse CYP1B1 expressed in Dermatol 99(5):42S–45S
Escherichia coli exhibits selective binding by polycyclic hydrocar- 90. Williams IR, Kupper TS (1996) Immunity at the surface: homeo-
bons and metabolism which parallels C3H10T1/2 cell microsomes, static mechanisms of the skin immune system. Life Sci
but differs from human recombinant CYP1B1. Arch Biochem 58(18):1485–1507
Biophys 347(2):181–192 91. Lai ZW, Pineau T, Esser C (1996) Identification of dioxin-
72. Wislocki PG, Gadek KM, Chou MW, Yang SK, Lu AY (1980) responsive elements (DREs) in the 5′ regions of putative dioxin-
Carcinogenicity and mutagenicity of the 3,4-dihydrodiols and other inducible genes. Chem Biol Interact 100(2):97–112
metabolites of 7,12-dimethylbenz(a)anthracene and its 92. Henley DV, Bellone CJ, Williams DA, Ruh MF (2004) MAPK
hydroxymethyl derivatives. Cancer Res 40(10):3661–3664 signaling pathways modulate IL-1beta expression in human
73. Tsuchiya Y, Nakajima M, Kyo S, Kanaya T, Inoue M, Yokoi T keratinocytes. Arch Biochem Biophys 424(1):112–118
(2004) Human CYP1B1 is regulated by estradiol via estrogen 93. Tsuji G, Takahara M, Uchi H, Takeuchi S, Mitoma C, Moroi Y et al
receptor. Cancer Res 64(9):3119–3125 (2011) An environmental contaminant, benzo(a)pyrene, induces
74. Zheng W, Jefcoate CR (2005) Steroidogenic factor-1 interacts with oxidative stress-mediated interleukin-8 production in human
cAMP response element-binding protein to mediate cAMP stimu- keratinocytes via the aryl hydrocarbon receptor signaling pathway.
lation of CYP1B1 via a far upstream enhancer. Mol Pharmacol J Dermatol Sci 62(1):42–49
67(2):499–512 94. Carvajal-Gonzalez JM, Roman AC, Cerezo-Guisado MI, Rico-
75. Uno S, Dalton TP, Shertzer HG, Genter MB, Warshawsky D, Leo EM, Martin-Partido G, Fernandez-Salguero PM (2009)
Talaska G et al (2001) Benzo[a]pyrene-induced toxicity: paradoxi- Loss of dioxin-receptor expression accelerates wound healing
cal protection in Cyp1a1(−/−) knockout mice having increased in vivo by a mechanism involving TGF{beta}. J Cell Sci 122(Pt 11):
hepatic BaP-DNA adduct levels. Biochem Biophys Res Commun 1823–1833
289(5):1049–1056 95. Kissenpfennig A, Henri S, Dubois B, Laplace-Builhe C, Perrin P,
76. Haarmann-Stemmann T, Abel J, Fritsche E, Krutmann J (2012) The Romani N et al (2005) Dynamics and function of Langerhans cells
AhR-Nrf2 pathway in keratinocytes: on the road to chemopreven- in vivo: dermal dendritic cells colonize lymph node areas distinct
tion? J Invest Dermatol 132(1):7–9 from slower migrating Langerhans cells. Immunity 22(5):643–654
77. Baird L, Dinkova-Kostova AT (2011) The cytoprotective role of the 96. Bennett CL, van Rijn E, Jung S, Inaba K, Steinman RM,
Keap1-Nrf2 pathway. Arch Toxicol 85(4):241–272 Kapsenberg ML et al (2005) Inducible ablation of mouse
78. Xu C, Huang MT, Shen G, Yuan X, Lin W, Khor TO et al (2006) Langerhans cells diminishes but fails to abrogate contact hypersen-
Inhibition of 7,12-Dimethylbenz(a)anthracene-induced skin tumor- sitivity. J Cell Biol 169(4):569–576
igenesis in C57BL/6 mice by sulforaphane is mediated by nuclear 97. Zhao X, Deak E, Soderberg K, Linehan M, Spezzano D, Zhu J et al
factor E2-related factor 2. Cancer Res 66(16):8293–8296 (2003) Vaginal submucosal dendritic cells, but not Langerhans cells,
79. Rundhaug JE, Fischer SM (2008) Cyclo-oxygenase-2 plays a crit- induce protective Th1 responses to herpes simplex virus-2. J Exp
ical role in UV-induced skin carcinogenesis. Photochem Photobiol Med 197(2):153–162
84(2):322–329 98. Allan RS, Smith CM, Belz GT, van Lint AL, Wakim LM, Heath
80. Ciolino HP, Daschner PJ, Yeh GC (1998) Resveratrol inhibits WR et al (2003) Epidermal viral immunity induced by CD8alpha +
transcription of CYP1A1 in vitro by preventing activation of the dendritic cells but not by Langerhans cells. Science 301(5641):
aryl hydrocarbon receptor. Cancer Res 58(24):5707–5712 1925–1928
81. Palermo CM, Hernando JI, Dertinger SD, Kende AS, Gasiewicz TA 99. Puhvel SM, Sakamoto M, Reisner RM (1989) Effect of TCDD on
(2003) Identification of potential aryl hydrocarbon receptor antago- the density of Langerhans cells in murine skin. Toxicol Appl
nists in green tea. Chem Res Toxicol 16(7):865–872 Pharmacol 99(1):72–80
82. Aziz MH, Reagan-Shaw S, Wu J, Longley BJ, Ahmad N (2005) 100. Kadow S, Jux B, Zahner SP, Wingerath B, Chmill S, Clausen BE
Chemoprevention of skin cancer by grape constituent resveratrol: et al (2011) Aryl hydrocarbon receptor is critical for homeostasis of
relevance to human disease? FASEB J 19(9):1193–1195 invariant gamma}{delta T cells in the murine epidermis. J Immunol
83. Lu YP, Lou YR, Xie JG, Peng QY, Liao J, Yang CS et al (2002) 187(6):3104–3110
Topical applications of caffeine or (−)-epigallocatechin gallate 101. Kadow S, Jux B, Chmill S, Esser C (2010) Small molecules as
(EGCG) inhibit carcinogenesis and selectively increase apoptosis friends and foes of the immune system. Future Med Chem 1(9):
in UVB-induced skin tumors in mice. Proc Natl Acad Sci U S A 1583–1591
99(19):12455–12460 102. Merk HF, Baron JM, Heise R, Fritsche E, Schroeder P, Abel J et al
84. Sun SY, Hail N Jr, Lotan R (2004) Apoptosis as a novel target for (2006) Concepts in molecular dermatotoxicology. Exp Dermatol
cancer chemoprevention. J Natl Cancer Inst 96(9):662–672 15(9):692–704
85. Rosette C, Karin M (1996) Ultraviolet light and osmotic stress: 103. Ito T, Inouye K, Nohara K, Tohyama C, Fujimaki H (2008) TCDD
activation of the JNK cascade through multiple growth factor and exposure exacerbates atopic dermatitis-related inflammation in NC/
cytokine receptors. Science 274(5290):1194–1197 Nga mice. Toxicol Lett 177(1):31–37
86. Tran QT, Kennedy LH, Leon CS, Bodreddigari S, Goodwin SB, 104. Vogel CF, Matsumura F (2009) A new cross-talk between the aryl
Sutter CH et al (2012) EGFR regulation of epidermal barrier func- hydrocarbon receptor and RelB, a member of the NF-kappaB fam-
tion. Physiol Genomics 44(8):455–469 ily. Biochem Pharmacol 77(4):734–745
87. Sutter CH, Yin H, Li Y, Mammen JS, Bodreddigari S, Stevens G 105. Kimura A, Naka T, Nakahama T, Chinen I, Masuda K, Nohara K
et al (2009) EGF receptor signaling blocks aryl hydrocarbon et al (2009) Aryl hydrocarbon receptor in combination with Stat1
690 Semin Immunopathol (2013) 35:677–691

regulates LPS-induced inflammatory responses. J Exp Med 125. Opitz CA, Litzenburger UM, Sahm F, Ott M, Tritschler I, Trump S
206(9):2027–2035 et al (2011) An endogenous tumor-promoting ligand of the human
106. Veldhoen M, Hirota K, Westendorf AM, Buer J, Dumoutier L, aryl hydrocarbon receptor. Nat Geosci 478(7368):197–203
Renauld JC et al (2008) The aryl hydrocarbon receptor links 126. Ma Q (2011) Influence of light on aryl hydrocarbon receptor
TH17-cell-mediated autoimmunity to environmental toxins. Nature signaling and consequences in drug metabolism, physiology,
453(7191):106–109 and disease. Expert Opin Drug Metab Toxicol 7(10):1267–
107. Quintana FJ, Basso AS, Iglesias AH, Korn T, Farez MF, Bettelli E 1293
et al (2008) Control of T(reg) and T(H)17 cell differentiation by the 127. Korkina LG, Pastore S, Dellambra E, De LC (2012) New molecular
aryl hydrocarbon receptor. Nature 453(7191):65–71 and cellular targets for chemoprevention and treatment of skin
108. Ito M, Ogawa K, Takeuchi K, Nakada A, Heishi M, Suto H et al tumors by plant polyphenols: a critical review. Curr Med Chem
(2004) Gene expression of enzymes for tryptophan degradation 128. Platzer B, Richter S, Kneidinger D, Waltenberger D, Woisetschlager
pathway is upregulated in the skin lesions of patients with atopic M, Strobl H (2009) Aryl hydrocarbon receptor activation inhibits
dermatitis or psoriasis. J Dermatol Sci 36(3):157–164 in vitro differentiation of human monocytes and Langerhans den-
109. Voorhis MV, Fechner JH, Zhang X, Mezrich JD (2012) The Aryl dritic cells. J Immunol 183(1):66–74
hydrocarbon receptor: a novel target for immunomodulation in 129. Sibilano R, Frossi B, Calvaruso M, Danelli L, Betto E, Dall’Agnese
organ transplantation. Transplantation A et al (2012) The aryl hydrocarbon receptor modulates acute and
110. Apetoh L, Quintana FJ, Pot C, Joller N, Xiao S, Kumar D et al late mast cell responses. J Immunol 189(1):120–127
(2010) The aryl hydrocarbon receptor interacts with c-Maf to pro- 130. Cho YC, Zheng W, Jefcoate CR (2004) Disruption of cell–cell
mote the differentiation of type 1 regulatory T cells induced by IL- contact maximally but transiently activates AhR-mediated transcrip-
27. Nat Immunol 11(9):854–861 tion in 10 T1/2 fibroblasts. Toxicol Appl Pharmacol 199(3):220–
111. Gandhi R, Kumar D, Burns EJ, Nadeau M, Dake B, Laroni A et al 238
(2010) Activation of the aryl hydrocarbon receptor induces human 131. Ju Q, Fimmel S, Hinz N, Stahlmann R, Xia L, Zouboulis CC (2011)
type 1 regulatory T cell-like and Foxp3(+) regulatory T cells. Nat 2,3,7,8-Tetrachlorodibenzo-p-dioxin alters sebaceous gland cell dif-
Immunol 11(9):846–853 ferentiation in vitro. Exp Dermatol 20(4):320–325
112. Fujita H, Nograles KE, Kikuchi T, Gonzalez J, Carucci JA, Krueger 132. Singh KP, Casado FL, Opanashuk LA, Gasiewicz TA (2009) The
JG (2009) Human Langerhans cells induce distinct IL-22-producing aryl hydrocarbon receptor has a normal function in the regulation of
CD4+ T cells lacking IL-17 production. Proc Natl Acad Sci U S A hematopoietic and other stem/progenitor cell populations. Biochem
106(51):21795–21800 Pharmacol 77(4):577–587
113. Trifari S, Kaplan CD, Tran EH, Crellin NK, Spits H (2009) Identi- 133. Esser C, Rannug A, Stockinger B (2009) The aryl hydrocarbon
fication of a human helper T cell population that has abundant receptor and immunity. Trends Immunol 9:447–454
production of interleukin 22 and is distinct from T(H)-17, T(H)1 134. Pauly SK, Fechner JH, Zhang X, Torrealba J, Bradfield CA,
and T(H)2 cells. Nat Immunol 10(8):864–871 Mezrich JD (2012) The aryl hydrocarbon receptor influences trans-
114. Zhang N, Pan HF, Ye DQ (2011) Th22 in inflammatory and auto- plant outcomes in response to environmental signals. Toxicol En-
immune disease: prospects for therapeutic intervention. Mol Cell viron Chem 94(6):1175–1187
Biochem 353(1–2):41–46 135. Schallreuter KU, Salem MA, Gibbons NC, Martinez A, Slominski
115. Wu HY, Quintana FJ, da Cunha AP, Dake BT, Koeglsperger T, R, Ludemann J et al (2012) Blunted epidermal L -tryptophan me-
Starossom SC et al (2011) In vivo induction of Tr1 cells via mucosal tabolism in vitiligo affects immune response and ROS scavenging
dendritic cells and AHR signaling. PLoS One 6(8):e23618 by Fenton chemistry, part 1: epidermal H2O2/ONOO(−)-mediated
116. Girardi M, Lewis JM, Filler RB, Hayday AC, Tigelaar RE (2006) stress abrogates tryptophan hydroxylase and Dopa decarboxylase
Environmentally responsive and reversible regulation of epidermal activities, leading to low serotonin and melatonin levels. FASEB J
barrier function by gamma delta T cells. J Invest Dermatol 26(6):2457–2470
126(4):808–814 136. Kramer HJ, Podobinska M, Bartsch A, Battmann A, Thoma W,
117. Martin B, Hirota K, Cua DJ, Stockinger B, Veldhoen M (2009) Bernd A et al (2005) Malassezin, a novel agonist of the aryl
Interleukin-17-producing gamma delta T cells selectively expand in hydrocarbon receptor from the yeast Malassezia furfur, induces
response to pathogen products and environmental signals. Immunity apoptosis in primary human melanocytes. Chembiochem
31(2):321–330 6(5):860–865
118. Li Y, Innocentin S, Withers DR, Roberts NA, Gallagher AR, 137. Gaitanis G, Magiatis P, Stathopoulou K, Bassukas ID, Alexopoulos
Grigorieva EF et al (2011) Exogenous stimuli maintain EC, Velegraki A et al (2008) AhR ligands, malassezin, and
intraepithelial lymphocytes via aryl hydrocarbon receptor activa- indolo[3,2-b]carbazole are selectively produced by Malassezia
tion. Cell. doi:10.1016/j.cell.2011.09.025 furfur strains isolated from seborrheic dermatitis. J Invest Dermatol
119. Ebert LM, Meuter S, Moser B (2006) Homing and function of 128(7):1620–1625
human skin gamma delta T cells and NK cells: relevance for tumor 138. Gaido KW, Maness SC, Leonard LS, Greenlee WF (1992) 2,3,7,8-
surveillance. J Immunol 176(7):4331–4336 Tetrachlorodibenzo-p-dioxin-dependent regulation of transforming
120. Jameson J, Havran WL (2007) Skin gamma-delta T cell functions in growth factors-alpha and -beta 2 expression in a human keratinocyte
homeostasis and wound healing. Immunol Rev 215:114–122 cell line involves both transcriptional and post-transcriptional con-
121. Holtmeier W, Kabelitz D (2005) Gamma-delta T cells link innate trol. J Biol Chem 267(34):24591–24595
and adaptive immune responses. Chem Immunol Allergy 86:151– 139. Loertscher JA, Lin TM, Peterson RE, Allen-Hoffmann BL (2002)
183 In utero exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin causes
122. Laggner U, Di MP, Perera GK, Hundhausen C, Lacy KE, Ali N et al accelerated terminal differentiation in fetal mouse skin. Toxicol
(2011) Identification of a novel proinflammatory human skin- Sci 68(2):465–472
homing Vgamma9Vdelta2 T cell subset with a potential role in 140. Sutter CH, Bodreddigari S, Campion C, Wible RS, Sutter TR (2011)
psoriasis. J Immunol 187(5):2783–2793 2,3,7,8-Tetrachlorodibenzo-p-dioxin increases the expression of
123. Cai Y, Fleming C, Yan J (2012) New insights of T cells in the genes in the human epidermal differentiation complex and acceler-
pathogenesis of psoriasis. Cell Mol Immunol 9(4):302–309 ates epidermal barrier formation. Toxicol Sci 124(1):128–137
124. Prud’homme GJ (2012) Cancer stem cells and novel targets for 141. Loertscher JA, Sattler CA, Allen-Hoffmann BL (2001) 2,3,7,8-
antitumor strategies. Curr Pharm Des 18(19):2838–2849 Tetrachlorodibenzo-p-dioxin alters the differentiation pattern of
Semin Immunopathol (2013) 35:677–691 691

human keratinocytes in organotypic culture. Toxicol Appl events as risk factors for psoriasis: results from an Italian case–
Pharmacol 175(2):121–129 control study. J Invest Dermatol 125(1):61–67
142. Villano CM, Murphy KA, Akintobi A, White LA (2006) 2,3,7,8- 144. D’Cruz D (2000) Autoimmune diseases associated with drugs,
tetrachlorodibenzo-p-dioxin (TCDD) induces matrix metallopro- chemicals and environmental factors. Toxicol Lett 112–113:421–432
teinase (MMP) expression and invasion in A2058 melanoma cells. 145. Rieber N, Belohradsky BH (2010) AHR activation by tryptophan-
Toxicol Appl Pharmacol 210(3):212–224 pathogenic hallmark of Th17-mediated inflammation in eosinophil-
143. Naldi L, Chatenoud L, Linder D, Belloni FA, Peserico A, Virgili AR ic fasciitis, eosinophilia-myalgia-syndrome, and toxic oil syn-
et al (2005) Cigarette smoking, body mass index, and stressful life drome? Immunol Lett 128(2):154–155
Copyright of Seminars in Immunopathology is the property of Springer Science & Business
Media B.V. and its content may not be copied or emailed to multiple sites or posted to a
listserv without the copyright holder's express written permission. However, users may print,
download, or email articles for individual use.

You might also like