Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Feb 21.
Published in final edited form as: Nat Rev Immunol. 2012 Jan 13;12(2):114–124. doi: 10.1038/nri3150

Early events in the induction of allergic contact dermatitis

Daniel H Kaplan 1, Botond Z Igyártó 1, Anthony A Gaspari 2
PMCID: PMC3578582  NIHMSID: NIHMS433436  PMID: 22240625

Preface

The skin is a barrier site that is exposed to a wide variety of potential pathogens. As in other organs, pathogens that invade the skin are recognized by pattern-recognition receptors. Recently, it has been appreciated that pattern-recognition receptors are also engaged by chemical contact allergens. This elicits an inappropriate immune response in susceptible individuals resulting in allergic contact dermatitis. In this Review, we will focus on how contact allergens promote inflammation. We will also examine how immune cells in the skin, including mast cells, T cells and dendritic cells, cooperate with each other and with keratinocytes to drive early responses to contact allergens.

Introduction

Allergic contact dermatitis is a common skin disease that is caused by type IV delayed type hypersensitivity responses to antigens that come in contact with the skin (Box 1)1. Allergic contact dermatitis is a major cause of occupational skin disease, and accounts for approximately 20% of all work-related health complaints. It results in an estimated 4 million lost work-days and has an associated cost of almost $400 million per year in the United States alone2-4. One of the clinically most important categories of contact allergens is small organic molecules that are chemically reactive (chemical sensitizers). They bind with self proteins to generate immunogenic neo-antigens through a process termed haptenization. Contact allergens are common in cosmetics, personal care products, jewelry and in the workplace whether it is in an industrial, health care, or office setting. Examples of common allergens are inorganic chemicals such a nickel and organic chemicals such as those found in fragrances and dyes.

Box 1. Allergic contact dermatitis versus contact hypersensitivity.

Allergic contact dermatitis is a clinical term used to describe an inflammatory reaction in the skin. Allergic contact dermatitis is characterized by a wide range of symptoms and patients are typically described as having an inflammatory reaction (dermatitis) with intensely pruritic erythema, edema and even vesicles at sites where chemicals contact the skin. In humans, allergic contact dermatitis usually occurs in response to low molecular weight compounds after many repeated sub-threshold exposures that do not initially induce allergic signs and symptoms. This is termed the ‘afferent’ or ‘sensitizing’ phase of allergic contact dermatitis. The afferent phase may take weeks to months of repeated exposures to the sensitizing chemical. Gradually, the patient becomes allergic to the offending compound and dermatitis occurs on further exposures to the allergen. This is the ‘efferent’ or ‘elicitation’ phase of allergic contact dermatitis. There are over 2800 chemicals that have the potential to cause allergic contact dermatitis 113. Patch testing is a standardized, diagnostic procedure that is necessary to confirm allergic contact dermatitis. Potential allergens are applied onto a patient’s back skin. After 2 days, the site of application of each potential allergen is evaluated. A positive patch test recapitulates the efferent phase of allergic contact dermatitis (that is, the allergen induces dermatitis) and this helps to identify the initial offending chemical. The treatment of choice for allergic contact dermatitis is strict allergen avoidance of the offending chemical. Pharmacologic measures, such as topical and systemic glucocorticosteroids and oral antihistamines are also frequently used to control allergic contact dermatitis.

Contact hypersensitivity is an experimental model system for human allergic contact dermatitis. Typically, this model utilizes rodents, such as mice. Rather than a series of sub-clinical exposures as occurs in humans, potent lipophilic compounds (such as dinitrofluorobenzene, and other similar chemicals) are dissolved in an organic solvent and applied directly to the shaved abdominal skin of the experimental animal over a period of 1 or 2 days (this is the afferent phase of contact hypersensitivity). After five to seven days, an elicitation response is induced by application of the same compound to the ear or footpad. The amount of swelling that occurs in the ear or footpad correlates with the intensity of the effector response generated. Contact hypersensitivity depends on the development of an early inflammatory response in the skin that is driven by innate mechanisms, but ultimately requires antigen-specific CD8+ cytotoxic T lymphocytes (CTLs) and CD4+ helper T cells. Contact hypersensitivity is considered an example of a type IV hypersensitivity reaction (T cell mediated allergic reaction). Although the circumstances of sensitization differ between humans with allergic contact dermatitis and mice with contact hypersensitivity, the contact hypersensitivity model is considered a reliable model for allergic contact dermatitis and has provided the framework for understanding this disease.

There are a series of fundamental events that lead to immune recognition of the hapten–self complex (Box 2). The first is percutaneous penetration through the stratum corneum barrier, which is the water impermeable outer layer of skin. Compounds must be less than 500 Daltons for efficient penetration5. This step allows the chemical sensitizer to come into contact and haptenize self-proteins in the skin. Haptens or haptenated self proteins are recognized by innate immune mechanisms in the skin and this leads to the elaboration of a number of pro-inflammatory mediators, including interleukin-1β (IL-1β). As a result, skin-resident dendritic cells (DCs) become activated. These DCs, which may have been directly haptenated or could have acquired haptenated proteins from their surroundings, migrate to skin-draining lymph nodes where they present peptides from haptenated proteins to activate memory and naïve T cells. In the final step, hapten-induced inflammation recruits activated effector T cells back to the initial site of antigen encounter in the skin (Box 3). The effector T cells release pro-inflammatory cytokines, such as IFNγ, and promote the killing of haptenized cells resulting in the development of the classic inflammatory rash that is seen in patients with allergic contact dermatits5-8.

Box 2. Essential steps for induction of ACD.

Percutaneous penetration of hapten

Haptenization of self proteins

Epidermal and dermal-derived inflammation

Dendritic cell activation and migration from skin to local lymph node

Box 3. Development of the effector response.

Allergic contact dermatitis can be considered a cascade of sequential and parallel events that starts with the innate immune system sensing self-damage that occurs during the haptenization process. This generates an inflammatory response that mobilizes cutaneous DCs to mature, migrate and traffic haptenated self proteins to local lymph nodes for presentation to antigen-specific T cells. As a consequence, naïve CD4+ and CD8+ T cells differentiate in the lymph node into effector T 114 H1, TH17 and CTL cells . These cells are then recruited back to the skin115 where CTL mediate apoptosis of keratonicytes116. In addition to effectors, regulatory CD4+ Treg develop that inhibit ACD and mediate tolerance in nonallergic individuals117, 118. NK cells in mice have been shown capable of maintaining persistent, hapten-specific memory119. Although these studies have not been translated to humans, NK cells are present in significant numbers in cellular infiltrates of patients with allergic contact dermatitis120, 121. Invariant NKT cells are thought to respond to endogenous glycolipids and secrete cytokines such as IFNγ that help amplify the early innate response122. Defective NKT cells or application of antagonist lipids that inhibit NKT cell activation results in reduced contact hypersensitivity in mice123, 124.

In this Review, we will first examine how the hapten–self complex and other contact allergens are recognized by the innate immune system. We will then focus on each of the skin-resident immune cells that contribute to the allergen-induced inflammatory cascade and how they ultimately lead to the generation of the adaptive response that results in allergic contact dermatitis.

Hapten–self complex formation

The landmark article of Landsteiner and Jacobs published in 1935 on the sensitizing potential of 2,4-dinitrohalobenzene derivatives led to the hypothesis that the ability of a chemical to react covalently with a ‘carrier protein’ is a major determinant factor in its ability to act as a skin sensitizer9-11. Indeed, the formation of the hapten–carrier complex in which the xenobiotic chemical attaches itself to host molecule(s) generates a neo-antigen that is eventually recognized by the immune system as ‘altered self’. The most reactive amino acid nucleophilic side-chains are those that are found on lysine, cysteine and histidine residues. However, their degree of ionization, and hence nucleophilicity is dependent on the pH of the microenvironment, which is influenced by surrounding amino acids as well as by protein location within the epithelium12.

The ability of the chemical species to react covalently with the carrier protein is related to electrophilic reactivity and hydrophobicity. Complete haptens are chemicals with skin sensitization potential that are directly reactive (electrophilic) with protein nucleophiles. Pro-haptens, which are also hydrophobic molecules, require activation by the host metabolism to make them into a reactive electrophile intermediary that can conjugate with host proteins (Figure 1)13. A large number of enzymes have been suggested to participate in activation of pro-haptens, including the cytochrome P450 (P450) mixed-function oxidase system, alcohol dehydrogenases, aldehyde dehydrogenases , monoamine oxidases , flavin-containing monooxygenases , hydrolytic enzymes, acyltransferases, glutathione S-transferases, uridine 5′-diphospho - glucuronosyltransferases and sulfotransferases14-16.

Pre-haptens are chemicals that are not activated by host proteins but instead require chemical transformation by oxidative derivatization by ambient or air oxidation to form hydroperoxides. Common examples include urushiol, which is derived from the poison ivy plant, certain fragrance materials and dyes used in hair coloring, such as para-phenylene diamine17. Finally, metal salts such as nickel interact with amino acids such as histidine to form non-covalent chelation complexes.

Allergen recognition

Innate recognition of haptens

It has long been recognized that the presence of foreign antigen alone is insufficient to generate immune responses: activation of the innate immune system is also required. In the case of vaccines, this is accomplished by combining antigen with adjuvants. In most cases, adjuvants contain microbial products (for example, mycobacteria in complete Freund’s adjuvant) that engage families of receptors which have evolved to recognize specific components of microbial organisms, that is, pathogen-associated molecular patterns (PAMPs). Many of these receptors can also respond to endogenous ligands to sense cell damage or necrosis. Historically, research into the immunology of acute contact dermatitis and contact hypersensitivity has overlooked hapten-mediated activation of the innate immune system to focus on the role of cutaneous antigen-presenting cells (APCs), such as Langerhans cells and dermal DC, and conventional CD8+ and CD4+ T cells. The importance of hapten-mediated activation of innate immunity is highlighted by the clinical observation that the irritancy of chemicals (that is, the ability of these chemicals to cause grossly visible skin inflammation upon primary exposure) correlates with their ability to act as contact sensitizers and to induce acute contact dermatitis.

Haptens activate Toll-like receptors

Toll-like receptors (TLRs) are a family of receptors that recognize PAMPs expressed by bacteria, parasites, viruses and fungi. TLRs are evolutionarily conserved in vertebrates and recognize a wide variety of ligands, including lipids, lipoproteins, proteins and nucleic acids18. TLRs trigger specific biological responses. For instance, TLR4-mediated recognition of lipopolysaccharide (LPS), a cell wall component found in gram-negative bacteria, induces the secretion of type I interferons (IFNs) and other pro-inflammatory cytokines. In contrast, activation of a cell via other TLRs, such as heterodimers of TLR1–TLR2 and TLR2–TLR6 or TLR5 homodimers, induces the secretion of pro-inflammatory cytokines but not IFN production. These differences are explained by differences in the downstream signaling pathways engaged by different TLRs. Most TLRs, with the exception of TLR3 and TLR4, signal exclusively via the adapter MyD88, which activates the transcription factor nuclear factor-κB (NF-κB) and mitogen-activated protein kinases (MAPKs) to induce the expression of pro-inflammatory cytokines, such as IL-1β18. TLR3 signals via TIR-domain-containing adaptor protein inducing IFNβ (TRIF) leading to activation of the transcription factors interferon-regulatory factor 3 (IRF3) and NF-κB and results in induction of type I IFN and inflammatory cytokines. TLR4 signals via both TRIF and MyD88 resulting in activation of both signalling pathways. In addition to PAMPs, TLRs also recognize damage associated molecular patterns (DAMPs), which are self molecules that can be released during necrotic cell death. PAMPs comprise a diverse set of proteins, nucleic acids, and glycosaminoglycans; examples include heat shock proteins, HMGB1, RNA, DNA, hyaluronan, and heparin sulfate19.

Recently, a crucial role for TLR2 and TLR4 has been described in contact hypersensitivity, an experimental model of allergic contact dermatitis (Box 1)20. Although mice deficient in either TLR2 or TLR4 developed normal contact hypersensitivity responses, mice lacking both of these TLRs were not sensitized and failed to develop contact hypersensitivity in response to the haptens trinitrochlorobenzene (TNCB), oxazolone and fluorescein isothiocyanate (FITC). Interestingly, if the mice were unable to respond to IL-12 the absence of either TLR2 or TLR4 alone was sufficient to prevent the development of optimal contact hypersensitivity20. These data suggest that there are two possible mechanisms of sensitization to contact allergens: an IL-12-dependent one, in which activation of TLR2 or TLR4 alone is sufficient for contact hypersensitivity to develop, and a IL-12-independent mechanism that requires activation of both TLR2 and TLR4.

Since TLR2 and TLR4 do not directly recognize haptens, a role for commensal skin bacteria was evaluated by performing sensitization experiments in germ-free mice. These studies showed that contact hypersensitivity to TNCB develops normally in the absence of the bacterial flora20. Thus, TLR2 and TLR4 appear to recognize endogenous host-derived ligands that are present in the skin, rather than components of the commensal flora. One candidate is low molecular weight breakdown products of high molecular weight hyaluronic acid (Figure 1)21. Low molecular weight-hyaluronic acid derivatives activate DCs in vitro via TLR2 and TLR422, 23. In addition, reactive oxygen species that are produced in response to haptens can induce breakdown of hyaluronic acid24. Importantly, blockade of hyaluronic acid function in germ-free mice significantly reduces sensitization to haptens20, which confirms a role for degraded hyaluronic acid as an endogenous activator of TLR2 and TLR4-mediated signaling in contact hypersensitivity responses. This observation has therapeutic implications, in that blockade of the formation of DAMPs may represent a novel approach to prevent allergic contact dermatitis in a naïve host, or lessen allergic contact dermatitis in an allergic host.

Haptens activate innate immunity via NLR receptors

A second system of PAMP recognition involves the formation of an intracellular complex of proteins termed the inflammasome (Figure 1)25. The inflammasome complex includes proteins of the nucleotide-binding domain, leucine-rich repeat-containing (NLR) gene family that sense foreign PAMPs or endogenous ligands through an incompletely understood mechanism. Engagement of these receptors leads to signaling through the adaptor protein ASC (apoptosis associated speck-like protein containing a caspase recruitment domain) to activate pro-caspase 1, which cleaves pro-IL-1β and pro-IL-18 into active forms. Since one outcome of TLR signaling is increased expression of mRNA for IL-1β, the TLR and inflammasome systems can work coordinately to induce the release of pro-inflammatory cytokines (Table 1).

Table 1.

Comparison on common skin contact allergens.

Allergen Allergic response Mechanism(s) of
Recognition
Comment
Nickel Acute contact
dermatitis in
humans
Engages critical
histidine residues in
human TLR4. Does not
bind mouse TLR432
Most common
contact allergen.
Barrier disruption
increases rate of
sensitization36
House Dust mite (Der
p2, Der f2)
Acute contact
dermatitis in
humans
Homologues of MD-2,
LPS-binding protein that
complexes with TLR434
Important skin and
respiratory antigen
for patients with
atopic dermatitis
Dinitrofluorobenzene
(DNFB),
trinitrochlorobenzene
(TNCB).
Contact
hypersensitivity in
mice
Induce hyaluronic acid
degradation products
that activate TLR2 and
TLR4 and induction of
ATP release with
inflammasome
activation20, 29
Commonly used
experimental
haptens
Fluorescein
isothiocyanate (FITC)
Contact
hypersensitivity in
mice
Unknown Commonly used
vehicle (DBP)
induces TSLP that
promotes Th2 50
Dinitrothiocyanobenzene
(DNTB)
Contact
hypersensitivity in
mice
Weak hapten that does
not activate
inflammasome30
Can induce
tolerance to DNFB,
a structurally similar
hapten30
Dimethylbenzanthracene
(DMBA)
Contact
hypersensitivity in
mice
Unknown mode of
recognition.
In absence of TLR4,
contact hypersensitivity
is unaffected but the
Th1/Th17 cell balance
is altered125
Carcinogen and
contact allergen.

The importance of this system in responding to haptens is demonstrated by the reduced sensitization to TNCB and dinitrofluorobenzene (DNFB) that occurs in ASC−/−, NALP3−/−, and caspase1−/− mice26-28. At least in the case of TNCB and oxazalone, inflammasome activation also depends on the ATP receptor P2X 297. This indicates that inflammasome activation occurs indirectly as a result of cell damage and release of ATP. Interestingly, dinitrothiocyanobenzene (DNTB) is a weak sensitizer that can induce tolerance to the structurally related hapten DNFB. DNTB does not induce inflammasome-mediated secretion of active IL-1β and IL-1830. However, artificial inflammasome induction with sodium dodecyl sulfate (SDS) or co-administration of IL-1β converts DNTB into a sensitizing agent. In addition, administration of low, sub-threshold doses of many haptens including DNFB can induce a long-lasting tolerance31. Thus, inflammasome activation may be a feature common to all sensitizing haptens.

Other skin allergens engage PRRs

Although the above mechanisms appear to be broadly applicable to allergic contact dermatitis, there are very specific examples of how PRRs are activated by certain human skin allergens. Nickel is the most common cause of allergic contact dermatitis and is known to activate NF-kB and MAPK signaling. Recently, human TLR4 was shown to be the crucial receptor for nickel-induced pro-inflammatory gene expression32. Nickel allergies in humans appear to be an accident of nature. Binding of nickel, but not of the natural ligand LPS, to human TLR4 requires the presence of two non-conserved histidine residues in human TLR4, H456 and H458. These histidines are absent from mouse TLR4, which explains the mystery of why mice fail to develop contact hypersensitivity to nickel32.

House dust mites can exacerbate eczema in patients with atopic dermatitis, and are also a major source of aeroallergens for patients with allergic asthma and activate innate immunity through a unique and important mechanism33. Concentrated in mite faecal pellets, the major group 2 allergens, Der p 2 and Der f 2, are highly allergenic; among defined dust mite antigens, they have the highest rates of skin test positivity in patients with atopic dermatitis, and can also result in exacerbations of eczema. Sequence homology places these allergens in the recently recognized MD2-related lipid-recognition domain family of proteins34. Der p 2 also has functional homology, facilitating signaling through direct interactions with the TLR4 complex, and reconstituting LPS-driven TLR4 signaling in the absence of MD-2. This property provides it with natural adjuvant properties that are highly relevant for its potency as a common skin and respiratory track allergen.

Cellular response to cutaneous allergen

In the preceding section, we have examined the molecular mechanisms underlying recognition of haptens. In this section we will examine how the different cells types in the skin contribute to the generation of adaptive hapten-specific immune responses. The skin can be broadly divided into two distinct regions, the epidermis and dermis (Figure 2). The epidermis is derived from ectoderm and is an epithelial layer composed primarily of keratinocytes that provide for the integrity of the skin. Langerhans cells and dendritic epidermal T cells (DETCs) reside within the epidermis. The dermis is a relatively cell-sparse tissue derived from mesoderm that forms a stromal layer immediately below the epidermis. It is populated by fibroblasts that secrete components of the complex extracellular matrix. In the steady-state, a variety of immune cells can be found in the dermis including T cells, mast cells, and dermal DCs.

Keratinocytes

Keratinocytes are a crucial cell type for the development of allergic contact dermatitis due to their numerical abundance in the epidermis and through their role in the formation of the anatomic barrier function of the skin. Although haptens can penetrate through intact skin, the importance of an intact skin barrier in limiting sensitivity to allergic contact dermatitis has long been hypothesized based on the observation that patients with certain disease states that impair barrier function (for example, leg ulcers and peri-anal dermatitis) have an increased risk of sensitization to topically applied medications and their vehicle components. Recently, direct evidence has been obtained through the discovery that many patients with atopic dermatitis (an inherited form of allergic chronic dermatitis) or allergic contact dermatitis to nickel harbor a defective form of the filaggrin gene35, 36. Filaggrin helps aggregate cytoskeletal proteins that form the cornified cell envelope. In its absence, the barrier is defective as determined by increased loss of water through the epidermis. The importance of filaggrin has been confirmed in mice into which the mutations observed in atopic dermatitis patients were genetically introduced37. Mice with defective filaggrindevelop exaggerated responses to percutaneous immunization with proteins and haptens that results in a reduced threshold for allergic dermatitis37-39.

In addition to physically blocking penetration, keratinocytes also recognize haptens via the mechanisms discussed above to generate the innate responses that are required for contact hypersensitivity (Figure 3a). Keratinocytes express most TLRs, except TLR7 and TLR8, and this allows them to respond to TLR4-triggering haptens, such as DNFB and nickel40. Inflammasome-mediated secretion of IL-1β and IL-18 occurs in response to certain haptens. In addition to IL-1β and IL-18, keratinocytes secrete a large number of other pro-inflammatory cytokines and chemokines (reviewed in 41, 42). Of particular importance is TNFα which, along with IL-1β and IL-18, is required for hapten-induced DC maturation and migration from the skin to the local lymph node43-45. TNFα may be partially redundant, though, as TNFα−/− mice have only somewhat reduced CHS46.

It has been recently appreciated that in addition to inducing DC migration, keratinocytes can modulate the T cell response through ‘conditioning’ of skin-resident DCs. Perhaps the best example of this is the conditioning effects of keratinocyte-derived thymic stromal lymphopoietin (TSLP) on DCs47. TSLP activates DCs to induce naive T cell differentiation into T helper 2 (TH2) cells, which promote allergic responses48, 49. The expression of TSLP is induced by dibutylphthalate (DBP), a vehicle often used in conjunction with FITC to generate a TH2-type contact hypersensitivity response. The importance of TSLP has been confirmed with the observation that mice deficient in the TSLP receptor develop reduced contact hypersensitivity responses to FITC–DBP50. TSLP is also expressed by keratinocytes isolated from patients with atopic dermatitis and may be a key contributor to the disease48. A second conditioning factor, the receptor activator of –NF-κB ligand (RANKL), is expressed by keratinocytes in response to inflammation. Overexpression of RANKL by keratinocytes increases the number of regulatory T (TReg) cells, most likely through effects on Langerhans cells that express RANK51. Interestingly, UV-light and vitamin D3, which both increase expression of RANKL by keratinocytes, suppress contact hypersensitivity responses in mice and allergic contact dermatitis in humans52-54.

In addition to promoting TReg cell responses through RANKL expression, keratinocytes also participate in suppressing the response. For example, the anti-microbial peptide, cathelicidin, is secreted by keratinocytes and inhibits hyaluronan-induced cytokine release55. Cathelicidin-deficient mice develop exaggerated contact hypersensitivity to DNFB. Activated keratinocytes express high levels of MHC class I and MHC class II but have only low levels of expression of the costimulatory molecule CD80 (also known as B7.1). Transgenic overexpression of CD80 on keratinocytes leads to exaggerated contact hypersensitivity, which suggests that the absence of CD80 on keratinocytes could inhibit or anergize effector T cells56, 57. Keratinocytes are also a source of IL-10, an immunosuppressive cytokine that limits the extent of contact hypersensitivity58.

Dendritic cells

The key event in allergic contact dermatitis is the priming of hapten-specific naïve CD4+ and CD8+ T cells, which then become activated, proliferate, and differentiate into effector T cell subsets. This occurs in the regional lymph node. Although haptens can drain to the lymph node via lymphatic flow this does not lead to a productive T cell response59, 60. Instead, antigen presentation in the lymph node by migratory DCs is absolutely essential for the generation of responses to peripheral antigen59-61,62.

In the skin at steady state, there are several populations of DCs. Langerhans cells are the only DC subtype in the epidermis. Langerhans cell precursors first populate the epidermis on embryonic day 18 and proliferate in situ to form a radio-resistant, self-renewing population that is unique among dendritic cells (DCs)63, 64. Like all skin-resident DCs, Langerhans cells efficiently acquire antigen in the periphery and migrate to regional lymph nodes where they present antigen to naïve and memory T cells65. Hapten also penetrates into the dermis and is acquired by dermal DCs66. Langerin+ dermal DCs (also referred to as CD103+ dermal DCs) are a distinct DC subset and arise from bone marrow precursors in a FMS-related tyrosine kinase 3 (FLT3)-dependent manner67-71. Although they represent only a small percentage of the total DC population in the dermis (~3%), Langerin+ dermal DCs are in a high state of flux and are continually replaced by new recruits from the blood68, 72. Examination of subset ontogeny has clearly identified that Langerhans cells and Langerin+ dermal DCs are distinct DC subsets63, 71, 73, 74 The second major dermal population, Langerin dermal DCs (also referred to as classic dermal DCs), make up the bulk of dermal DCs (~80%) and are heterogeneous, with at least two subsets that can be distinguished based on the expression of CD11b72. In response to inflammation, GR1+ monocytes are recruited to the skin where they develop into DC75, 76. The functional importance to CHS of this important DC subset is unclear.

Studies examining the specific contribution of steady-state DC subsets to the hapten-specific immune response have been made possible by the existence of several mouse strains that lack specific DC subsets (Table 2). HuLangerin-DTA mice express the active subunit of diphtheria toxin (DTA) specifically in Langerhans cells and this leads to a constitutive absence of epidermal Langerhans cells 77. HuLangerin-DTR mice express the primate diphtheria toxin receptor (DTR) in Langerhans cells and this enables inducible and specific ablation of Langerhans cells after injection of diphtheria toxin78. These mice develop enhanced contact hypersensitivity responses to various contact allergens, but not irritants such as benzalkonium chloride. Experiments in which Langerhans cells were depleted at different time points showed that Langerhans cells have suppressive functions during the priming phase but not during the effector phase of the contact hypersensitivity response77-79. Contact hypersensitivity responses are also exaggerated in huLangerin-Cre MHC-IIflox and huLangerin-Cre IL-10flox mice suggesting that Langerhans cells suppress contact hypersensitivity responses in an IL-10-dependent manner and require cognate interaction with CD4+ T cells (Figure 3b)79, 80. In addition to inhibiting contact hypersensitivity responses, Langerhans cells have suppressive functions in other immune models, and have been shown to promote tolerance to minor-mismatched skin grafts, suppress the clearance of Leishmania infection and mediate UV-induced immunosuppression 81-84.

Table 2.

Effects of genetic ablation of DC subsets on the development of contact hypersensitivity

Mouse strain Effects on dermal DC subsets Effect on
intensity of
contact
hypersensitivity
response
Epidermal
Langerhans
cells
Dermal
Langerin+
DCs
Dermal
Langerin-
DCs
CD8+
DCs
Constitutive systems
huLangerin-DTA77 absent normal normal normal graphic file with name nihms-433436-t0001.jpg
huLangerin-Cre79x IL-10flox
or huLangerin-Cre x I-Abflox
Functionally
defective
normal normal normal graphic file with name nihms-433436-t0002.jpg
Batf3−/−73 normal absent normal absent normal
Inducible systems
muLangerin-DTR (diphtheria
toxin administered day +1)85,
86, 91
absent absent normal ~25%
of DCs
present
graphic file with name nihms-433436-t0003.jpg
muLangerin-DTR (diphtheria
toxin administered day
++7/13)67, 91
absent ~20-50%
of DCs
present
normal normal normal
huLangerin-DTR
(diphtheria toxin administered
day +1)78
absent normal normal normal graphic file with name nihms-433436-t0004.jpg

As discussed above, Langerin is expressed in Langerhans cells and a subset of dermal DC. In mice that express DTA or DTR under control of the human Langerin promoter, dermal DCs are not deleted. This is presumably due to difference between the human and murine langerin promoters. In contrast, in muLangerin-DTR mice, diphtheria toxin injection ablates Langerhans cells, Langerin+ dermal DCs and also CD103+ DCs from many other peripheral tissues85, 86. CD8+ DCs that reside in lymph nodes and spleen are also eliminated with reduced efficiency. Contact hypersensitivity is greatly diminished in these mice if diphtheria toxin is administered prior to priming, but not prior to elicitation85-87. Langerhans cells repopulate the skin after diphtheria toxin-mediated ablation much more slowly than Langerin+ dermal DCs67. Interestingly, contact hypersensitivity responses develop normally in muLangerin-DTR mice if priming is delayed until 7-13 days after diphtheria toxin administration, a time-point when Langerhans cells are absent but Langerin+ dermal DCs have partially returned. This suggests that Langerin+ dermal DCs, rather than Langerhans cells, promote the development of contact hypersensitivity 67, 88. This is supported by the observation that contact hypersensitivity responses can be adoptively transferred with dermal DCs, but not with Langerhans cells, isolated from sensitized mice89. However, by using the delayed contact hypersensitivity technique or muLangerin-DTR bone marrow chimeras to selectively ablate Langerhans cells or Langerin+ dermal DCs, other groups have found that Langerin+ dermal DCs and Langerhans cells are functionally redundant90, 91. Thus, distinguishing the function of Langerhans cells from Langerin+ dermal DCs using muLangerin-DTR mice is complicated by the complex experimental manipulations that are required to deplete individual DC subsets. However, in Batf3−/− mice, which have a constitutive absence of Langerin+ dermal DCs73, contact hypersensitivity responses develop normally indicating that, at least in the setting of a constitutive absence of Langerin+ dermal DCs, these cells are not required for contact hypersensitivity.

A major obstacle to untangling the functions of skin-resident DCs during contact hypersensitivity is the high degree of experimental variability that is intrinsic to the contact hypersensitivity assay. In addition, it is difficult to efficiently examine hapten-specific responses. In the setting of skin infections, however, the functional distinction between skin DC subsets is clearer. Langerin+ dermal DCs isolated from skin-draining lymph nodes during recurrent Herpes simplex virus infection efficiently cross-prime antigen-specific CD8+ T cells in vitro92. In contrast, other skin-resident DCs and lymph node-resident DCs are much less efficient. Similarly, skin infection with Candida albicans induces efficient activation of CD8+ T cells in vivo in Langerin-DTA mice, which lack Langerhans cells, but CD8+ T cell activation is defective in infected Batf3−/− mice, which lack Langerin+ dermal DCs93. Moreover, Langerin+ dermal DCs are required for TH1 cell differentiation in response to C. albicans infection, whereas Langerhans cells are required for the development of TH17 cell responses. Thus, Langerin+ dermal DCs and Langerhans cells are clearly functionally distinct in the setting of skin infection. This may also be true for CHS but its demonstration requires the development of new tools to analyze hapten-specific responses.

Mast Cells

Mast cells are hematopoetically derived cells that reside long-term in barrier tissue sites, such as skin and gut. Mast cells express the high affinity receptor for IgE, FcεRI, and can be activated via antigen-mediated cross-linking of surface IgE (Figure 3a). In addition, mast cells express TLRs and respond to many microbial products94. In response to IgE-mediated activation, mast cells induce an immediate wheal and flare response (within minutes) by releasing pre-formed granules that contain a wide variety of immunologically active compounds, such as histamine, proteases, proteoglycans and TNF. Mast cells also secrete a number of pro-inflammatory ‘late’ mediators, such as IL-3, IL-4, IL-5, IL6, IL-8, IL-9, IL-11, IL-13, TNF, CCL3 (also known as MIP-1α), CCL4 (also known as MIP-1β) and CCL2 (also known as MCP1).

Though the participation of mast cells in the development of immune responses against pathogens has been well described (recently reviewed 95), the importance of mast cells during contact hypersensitivity is quite controversial. Early work found that mast cells were both redundant and required for contact hypersensitivity 96, 97. More recently, mast cell activation by hapten-specific IgM and complement has been shown to participate in the elicitation phase of contact hypersensitivity98. In addition, mast cell activation has been shown to induce migration of skin-resident DCs and to enhance the efficacy of immunizations99, 100. The availability of mice that lack mast cells due to mutations in the kit gene (that is, B6KitW-sh/W-sh or WBB6F1-KitW/Wv strains) has greatly facilitated evaluation of mast cell function in vivo101. These mast cell-deficient mice can be reconstituted with in vitro-derived mast cells derived from mice deficient in a particular cytokine, thereby functionally generating mice with a mast cell-specific cytokine deletion. Contact hypersensitivity responses to FITC are defective in B6KitW-sk/W-sh mice. Reconstitution of these mice with mast cells derived from wild-type mice, but not from TNF-deficient mice, restores contact hypersensitivity102. Similarly, contact hypersensitivity to oxazalone is defective in WBB6F1-KitW/Wv mice as well as in both IgE−/− and IgE receptor-deficient mice103. Thus, mast cells, mast cell activation via FcεRI, and mast cell-derived TNF are required for induction of contact hypersensitivity. Intriguingly, contact hypersensitivity to urushiol in B6KitW-sh/W-sh develops normally but fails to appropriately resolve resulting in increased ear swelling compared with wild-type mice several days after challenge. Reconstitution with wild-type, but not IL-10−/− mast cells, rescues the phenotype104. These experiments suggest that, at least in response to urushiol, mast cells also suppress the late phase of the contact hypersensitivity response by producing IL-10.

An important caveat to experiments using kit mutant mice is that these mice also have other defects. WBB6F1-KitW/Wv mice develop macrocystic anemia, have reduced numbers of neutrophils, lack melanocytes and are sterile 105. Though B6KitW-sh/W-sh mice have a milder phenotype, subtle effects outside the mast cell compartment cannot be excluded. Recently, transgenic mice have been engineered that have mast cell-specific expression of Cre recombinase (Mcpt5-Cre)106. Breeding Mcpt5-Cre mice with inducible-DTR mice, which ubiquitously express a loxP-stop-diphtheria toxin receptor construct, creates a mouse strain in which mast cells can be ablated by injection of diphtheria toxin 107. Mast cell ablation prior to sensitization or elicitation impairs contact hypersensitivity responses to FITC and oxazalone. In addition, Mcp5-Cre IL-10flox mice do not develop exaggerated contact hypersensitivity. Thus, mast cells are required for contact hypersensitivity, but mast cell-derived IL-10 does not appear to be necessary for the resolution of the contact hypersensitivity response. Interestingly, similar to earlier data using urushiol, late contact hypersensitivity responses to hapten are increased in B6KitW-sk/W-sh mice but this result is not due to the absence of mast cells, but rather from a previously unrecognized neutrophil defect in B6KitW-sk/W-sh mice.

γδ T cells

Dendritic epidermal T cells (DETC) that express the identical γδ TCR (Vγ5/Vδ 1 ) populate the epidermis of virtually all strains of inbred mice but are not found in other tissues. Their location in the epidermis brings them into contact with hapten during the sensitization stage of contact hypersensitivity. It is unknown whether they directly sense the presence of hapten, but they do respond to stress-induced molecules expressed by keratinocytes; for example, DETCs recognize RAE1 via NKG2D108. Several studies have suggested that these cells participate in contact hypersensitivity by modulating the response of αβ T cells109. However, TCR δ −/− mice, which lack all γδ T cells, develop normal contact hypersensitivity on a C57BL/6 genetic background. Interestingly, TCRδ −/− mice on either the FVB or NOD genetic backgrounds develop spontaneous dermatitis and enhanced contact hypersensitivity responses that can be rescued by the adoptive transfer of Vγ5/Vδ 1+ T cells, but not by transfer of other γδ T cellsthat do not express Vγ5/Vδ 1110. Due to a point mutation in a gene involved in the intrathymic positive selection of Vγ5/Vδ 1+ cells prior to their migration to the skin, the epidermis of the substrain of FVB mice produced by Taconic Farms (FVBTac mice) lacks Vγ5/Vδ 1+ DETCs and is instead populated by Vγ5/Vδ 1 DETCs. DETCs in FVBTac mice are functionally defective and these mice exhibit spontaneous dermatitis and develop exaggerated contact hypersensitivity responses that are similar to those seen in TCR δ −/− mice111. Thus, Vγ5/Vδ 1+ DETCs appear to suppress contact hypersensitivity through still poorly understood mechanisms. γδ T cells are also found in human skin and may be the functional equivalent of the rodent invariant epidermal γδ T cells112.

Conclusion

Understanding how contact allergens promote allergic contact dermatitis is important, not only for preventing this disease, but also to enable us to develop a better understanding of other inflammatory skin diseases. Keratinocytes, mast cells and skin-resident DCs all contribute to the recognition of contact allergens. The precise role of each cell type and the sequence of events is still being determined. For the small number of contact allergens studied so far, it appears that they are sensed by TLRs and the inflammasome. Since these recognition systems evolved to sense microbial pathogens, the inflammation induced by contact allergens appears to be an accident of nature. This raises the possibility that activation of these pathways is a trait shared by all compounds with sensitizing potential. This has important implications as determining whether specific compounds are sensitizers is required for ingredients that come in contact with human skin (for example, skin care products). Whether all 2800 of the currently known sensitizing chemicals also trigger inflammation via TLR and NRR pathways remains to be determined113.

Summary Bullet points.

  1. The ability of a chemical to react covalently with a ‘carrier protein’ is a major determinant factor in its ability to act as a skin sensitizer. The formation of the hapten–carrier complex generates a neo-antigen that is eventually recognized by the immune system as ‘altered self’.

  2. Haptens induce production of endogenous ligands for TLR and NLR pattern receptors that activate an innate immune response and is required for adaptive immune responses to hapten–carrier complex.

  3. Keratinocytes and mast cells produce inflammatory cytokines in response to hapten exposure that mobilizes skin-resident DC and recruits effector lymphocytes into the skin which are responsible for immune-mediated inflammatory response associated with ACD.

  4. Skin-resident dendritic cells are required for the development of an anti-hapten T cell response.

  5. The Langerhans cells subset of skin dendritic cells is not required for the generation of the anti-hapten response but the precise contribution of each dendritic cell subset is unclear.

Glossary

hapten

A molecule that can bind antibody but cannot by itself elicit an immune response. Antibodies that are specific for a hapten can be generated when the hapten is chemically linked to a protein carrier that can elicit a T-cell response.

dendritic epidermal T cells

(DETCs). γδ-TCR+ cells localized purely in the epidermis that are present in rodents and cattle, but not in humans. In mice, essentially all DETCs express precisely the same TCR, forming a prototype lymphocyte repertoire of limited diversity.

NKG2D

(Natural-killer group 2, member D). A lectin-type activating receptor that is encoded by the NK complex and is expressed at the surface of NK cells, NKT cells, γδT cells and some cytolytic CD8+ αβT cells. The ligands for NKG2D are MHC-class-I-polypeptide-related sequence A (MICA) and MICB in humans, and retinoic acid early transcript 1 (RAE1) and H60 in mice. Such ligands are generally expressed at the surface of infected, stressed or transformed cells.

Biographies

Gaspari Biography

Dr. Gaspari is currently the Shapiro Professor and Chairman of the Department of Dermatology at the University of Maryland Baltimore. He attended medical school at Jefferson Medical College, followed by a Dermatology residency at Emory Univeristy. Then, he performed his post-doctoral studies at the Dermatology Branch of the NCI in Bethesda, Maryland. His laboratory studies keratinocyte-lymphocyte interactions and how this regulates skin immunity. His laboratory also studies toll-like receptor signaling in the skin and how this influences responses to environmental challenges.

Kaplan Biography

Dr. Kaplan is currently the Zelickson Professor and Assistant Professor of Dermatology at the University of Minnesota, Minneapolis, MN. He completed his MD/PhD training at Washington University in St. Louis followed by training and fellowship at the Yale School of Medicine, New Haven, CT. His laboratory currently studies skin dendritic cells and how they contribute to the generation and regulation of cutaneous adaptive responses.

Igyarto Biography

Dr. Igyarto is currently a Resarch Associate in the department of Dermatology at the University of Minnesota, Minneapolis, MN. He completed his PhD training at Semmelweis University in Budapest. His work currently focuses on examining the mechanisms that determine why different skin DC subsets to promote distinct T-helper phenotypes.

References

  • 1.Mowad CM. Patch testing: pitfalls and performance. Curr Opin Allergy Clin Immunol. 2006;6:340–4. doi: 10.1097/01.all.0000244794.03239.8e. [DOI] [PubMed] [Google Scholar]
  • 2.Thyssen JP, Linneberg A, Menne T, Johansen JD. The epidemiology of contact allergy in the general population--prevalence and main findings. Contact Dermatitis. 2007;57:287–99. doi: 10.1111/j.1600-0536.2007.01220.x. [DOI] [PubMed] [Google Scholar]
  • 3.Diepgen TL. Occupational skin-disease data in Europe. Int Arch Occup Environ Health. 2003;76:331–8. doi: 10.1007/s00420-002-0418-1. [DOI] [PubMed] [Google Scholar]
  • 4.Diepgen TL, Coenraads PJ. The epidemiology of occupational contact dermatitis. Int Arch Occup Environ Health. 1999;72:496–506. doi: 10.1007/s004200050407. [DOI] [PubMed] [Google Scholar]
  • 5.Martin SF. T lymphocyte-mediated immune responses to chemical haptens and metal ions: implications for allergic and autoimmune disease. Int Arch Allergy Immunol. 2004;134:186–98. doi: 10.1159/000078765. [DOI] [PubMed] [Google Scholar]
  • 6.Vocanson M, et al. Contribution of CD4(+ )and CD8(+) T-cells in contact hypersensitivity and allergic contact dermatitis. Expert Rev Clin Immunol. 2005;1:75–86. doi: 10.1586/1744666X.1.1.75. [DOI] [PubMed] [Google Scholar]
  • 7.Martin SF, et al. Fas-mediated inhibition of CD4+ T cell priming results in dominance of type 1 CD8+ T cells in the immune response to the contact sensitizer trinitrophenyl. J Immunol. 2004;173:3178–85. doi: 10.4049/jimmunol.173.5.3178. [DOI] [PubMed] [Google Scholar]
  • 8.Watanabe H, Unger M, Tuvel B, Wang B, Sauder DN. Contact hypersensitivity: the mechanism of immune responses and T cell balance. J Interferon Cytokine Res. 2002;22:407–12. doi: 10.1089/10799900252952181. [DOI] [PubMed] [Google Scholar]
  • 9.Landsteiner K, Jacobs J. Studies on the Sensitization of Animals with Simple Chemical Compounds : Iii. Anaphylaxis Induced by Arsphenamine. J Exp Med. 1936;64:717–21. doi: 10.1084/jem.64.5.717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Aptula AO, Roberts DW. Mechanistic applicability domains for nonanimal-based prediction of toxicological end points: general principles and application to reactive toxicity. Chem Res Toxicol. 2006;19:1097–105. doi: 10.1021/tx0601004. [DOI] [PubMed] [Google Scholar]
  • 11.Roberts DW, Aptula AO. Determinants of skin sensitisation potential. J Appl Toxicol. 2008;28:377–87. doi: 10.1002/jat.1289. [DOI] [PubMed] [Google Scholar]
  • 12.Divkovic M, Pease CK, Gerberick GF, Basketter DA. Hapten-protein binding: from theory to practical application in the in vitro prediction of skin sensitization. Contact Dermatitis. 2005;53:189–200. doi: 10.1111/j.0105-1873.2005.00683.x. [DOI] [PubMed] [Google Scholar]
  • 13.Aptula AO, Roberts DW, Pease CK. Haptens, prohaptens and prehaptens, or electrophiles and proelectrophiles. Contact Dermatitis. 2007;56:54–6. doi: 10.1111/j.1600-0536.2007.00944.x. [DOI] [PubMed] [Google Scholar]
  • 14.Smith CK, Hotchkiss SAM. Metabolic Mechanisms. Taylor and Francis; London: 2001. pp. 119–205. [Google Scholar]
  • 15.Smith CK, Hotchkiss SAM. Allergic Contact Dermatitis: Chemical and Metabolic Mechanisms. Taylor and Francis; London: 2001. pp. 89–117. [Google Scholar]
  • 16.Kalgutkar AS, et al. A comprehensive listing of bioactivation pathways of organic functional groups. Curr Drug Metab. 2005;6:161–225. doi: 10.2174/1389200054021799. [DOI] [PubMed] [Google Scholar]
  • 17.Lepoittevin JP. Metabolism versus chemical transformation or pro-versus prehaptens? Contact Dermatitis. 2006;54:73–4. doi: 10.1111/j.0105-1873.2006.00795.x. [DOI] [PubMed] [Google Scholar]
  • 18.Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124:783–801. doi: 10.1016/j.cell.2006.02.015. [DOI] [PubMed] [Google Scholar]
  • 19.Sloane JA, Blitz D, Margolin Z, Vartanian T. A clear and present danger: endogenous ligands of Toll-like receptors. Neuromolecular Med. 12:149–63. doi: 10.1007/s12017-009-8094-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Martin SF, et al. Toll-like receptor and IL-12 signaling control susceptibility to contact hypersensitivity. J Exp Med. 2008;205:2151–62. doi: 10.1084/jem.20070509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Martin SF, et al. Mechanisms of chemical-induced innate immunity in allergic contact dermatitis. Allergy. 2011 doi: 10.1111/j.1398-9995.2011.02652.x. [DOI] [PubMed] [Google Scholar]
  • 22.Scheibner KA, et al. Hyaluronan fragments act as an endogenous danger signal by engaging TLR2. J Immunol. 2006;177:1272–81. doi: 10.4049/jimmunol.177.2.1272. [DOI] [PubMed] [Google Scholar]
  • 23.Termeer C, et al. Oligosaccharides of Hyaluronan activate dendritic cells via toll-like receptor 4. J Exp Med. 2002;195:99–111. doi: 10.1084/jem.20001858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Stern R, Kogan G, Jedrzejas MJ, Soltes L. The many ways to cleave hyaluronan. Biotechnol Adv. 2007;25:537–57. doi: 10.1016/j.biotechadv.2007.07.001. [DOI] [PubMed] [Google Scholar]
  • 25.Martinon F, Mayor A, Tschopp J. The inflammasomes: guardians of the body. Annu Rev Immunol. 2009;27:229–65. doi: 10.1146/annurev.immunol.021908.132715. [DOI] [PubMed] [Google Scholar]
  • 26.Sutterwala FS, et al. Critical role for NALP3/CIAS1/Cryopyrin in innate and adaptive immunity through its regulation of caspase-1. Immunity. 2006;24:317–27. doi: 10.1016/j.immuni.2006.02.004. [DOI] [PubMed] [Google Scholar]
  • 27.Watanabe H, et al. Activation of the IL-1beta-processing inflammasome is involved in contact hypersensitivity. J Invest Dermatol. 2007;127:1956–63. doi: 10.1038/sj.jid.5700819. [DOI] [PubMed] [Google Scholar]
  • 28.Antonopoulos C, et al. Functional caspase-1 is required for Langerhans cell migration and optimal contact sensitization in mice. J Immunol. 2001;166:3672–7. doi: 10.4049/jimmunol.166.6.3672. [DOI] [PubMed] [Google Scholar]
  • 29.Weber FC, et al. Lack of the purinergic receptor P2X(7) results in resistance to contact hypersensitivity. J Exp Med. 2010;207:2609–19. doi: 10.1084/jem.20092489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Watanabe H, et al. Danger signaling through the inflammasome acts as a master switch between tolerance and sensitization. J Immunol. 2008;180:5826–32. doi: 10.4049/jimmunol.180.9.5826. [DOI] [PubMed] [Google Scholar]
  • 31.Steinbrink K, Sorg C, Macher E. Low zone tolerance to contact allergens in mice: a functional role for CD8+ T helper type 2 cells. J Exp Med. 1996;183:759–68. doi: 10.1084/jem.183.3.759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Schmidt M, et al. Crucial role for human Toll-like receptor 4 in the development of contact allergy to nickel. Nat Immunol. 2010;11:814–9. doi: 10.1038/ni.1919. [DOI] [PubMed] [Google Scholar]
  • 33.Beltrani VS. The role of house dust mites and other aeroallergens in atopic dermatitis. Clin Dermatol. 2003;21:177–82. doi: 10.1016/s0738-081x(02)00366-8. [DOI] [PubMed] [Google Scholar]
  • 34.Trompette A, et al. Allergenicity resulting from functional mimicry of a Toll-like receptor complex protein. Nature. 2009;457:585–8. doi: 10.1038/nature07548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Palmer CN, et al. Common loss-of-function variants of the epidermal barrier protein filaggrin are a major predisposing factor for atopic dermatitis. Nat Genet. 2006;38:441–6. doi: 10.1038/ng1767. [DOI] [PubMed] [Google Scholar]
  • 36.Novak N, et al. Loss-of-function mutations in the filaggrin gene and allergic contact sensitization to nickel. J Invest Dermatol. 2008;128:1430–5. doi: 10.1038/sj.jid.5701190. [DOI] [PubMed] [Google Scholar]
  • 37.Fallon PG, et al. A homozygous frameshift mutation in the mouse Flg gene facilitates enhanced percutaneous allergen priming. Nat Genet. 2009;41:602–8. doi: 10.1038/ng.358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Oyoshi MK, Murphy GF, Geha RS. Filaggrin-deficient mice exhibit TH17-dominated skin inflammation and permissiveness to epicutaneous sensitization with protein antigen. J Allergy Clin Immunol. 2009;124:485–93. 493 e1. doi: 10.1016/j.jaci.2009.05.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Scharschmidt TC, et al. Filaggrin deficiency confers a paracellular barrier abnormality that reduces inflammatory thresholds to irritants and haptens. J Allergy Clin Immunol. 2009;124:496–506. 506 e1–6. doi: 10.1016/j.jaci.2009.06.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lebre MC, et al. Human keratinocytes express functional Toll-like receptor 3, 4, 5, and 9. J Invest Dermatol. 2007;127:331–41. doi: 10.1038/sj.jid.5700530. [DOI] [PubMed] [Google Scholar]
  • 41.Uchi H, Terao H, Koga T, Furue M. Cytokines and chemokines in the epidermis. J Dermatol Sci. 2000;24(Suppl 1):S29–38. doi: 10.1016/s0923-1811(00)00138-9. [DOI] [PubMed] [Google Scholar]
  • 42.Corsini E, Galli CL. Epidermal cytokines in experimental contact dermatitis. Toxicology. 2000;142:203–11. doi: 10.1016/s0300-483x(99)00145-6. [DOI] [PubMed] [Google Scholar]
  • 43.Nishibu A, Ward BR, Boes M, Takashima A. Roles for IL-1 and TNFalpha in dynamic behavioral responses of Langerhans cells to topical hapten application. J Dermatol Sci. 2007;45:23–30. doi: 10.1016/j.jdermsci.2006.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Cumberbatch M, Griffiths CE, Tucker SC, Dearman RJ, Kimber I. Tumour necrosis factor-alpha induces Langerhans cell migration in humans. Br J Dermatol. 1999;141:192–200. doi: 10.1046/j.1365-2133.1999.02964.x. [DOI] [PubMed] [Google Scholar]
  • 45.Cumberbatch M, Dearman RJ, Kimber I. Langerhans cells require signals from both tumour necrosis factor-alpha and interleukin-1 beta for migration. Immunology. 1997;92:388–95. doi: 10.1046/j.1365-2567.1997.00360.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Pasparakis M, Alexopoulou L, Episkopou V, Kollias G. Immune and inflammatory responses in TNF alpha-deficient mice: a critical requirement for TNF alpha in the formation of primary B cell follicles, follicular dendritic cell networks and germinal centers, and in the maturation of the humoral immune response. J Exp Med. 1996;184:1397–411. doi: 10.1084/jem.184.4.1397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Liu YJ, et al. TSLP: an epithelial cell cytokine that regulates T cell differentiation by conditioning dendritic cell maturation. Annu Rev Immunol. 2007;25:193–219. doi: 10.1146/annurev.immunol.25.022106.141718. [DOI] [PubMed] [Google Scholar]
  • 48.Soumelis V, et al. Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP. Nat Immunol. 2002;3:673–80. doi: 10.1038/ni805. [DOI] [PubMed] [Google Scholar]
  • 49.Ebner S, et al. Thymic stromal lymphopoietin converts human epidermal Langerhans cells into antigen-presenting cells that induce proallergic T cells. J Allergy Clin Immunol. 2007;119:982–90. doi: 10.1016/j.jaci.2007.01.003. [DOI] [PubMed] [Google Scholar]
  • 50.Larson RP, et al. Dibutyl phthalate-induced thymic stromal lymphopoietin is required for Th2 contact hypersensitivity responses. J Immunol. 2010;184:2974–84. doi: 10.4049/jimmunol.0803478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Loser K, et al. Epidermal RANKL controls regulatory T-cell numbers via activation of dendritic cells. Nat Med. 2006;12:1372–9. doi: 10.1038/nm1518. [DOI] [PubMed] [Google Scholar]
  • 52.Loser K, Beissert S. Regulation of cutaneous immunity by the environment: an important role for UV irradiation and vitamin D. Int Immunopharmacol. 2009;9:587–9. doi: 10.1016/j.intimp.2009.01.024. [DOI] [PubMed] [Google Scholar]
  • 53.Ghoreishi M, et al. Expansion of antigen-specific regulatory T cells with the topical vitamin d analog calcipotriol. J Immunol. 2009;182:6071–8. doi: 10.4049/jimmunol.0804064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Hanneman KK, Scull HM, Cooper KD, Baron ED. Effect of topical vitamin D analogue on in vivo contact sensitization. Arch Dermatol. 2006;142:1332–4. doi: 10.1001/archderm.142.10.1332. [DOI] [PubMed] [Google Scholar]
  • 55.Morioka Y, Yamasaki K, Leung D, Gallo RL. Cathelicidin antimicrobial peptides inhibit hyaluronan-induced cytokine release and modulate chronic allergic dermatitis. J Immunol. 2008;181:3915–22. doi: 10.4049/jimmunol.181.6.3915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Nasir A, Ferbel B, Salminen W, Barth RK, Gaspari AA. Exaggerated and persistent cutaneous delayed-type hypersensitivity in transgenic mice whose epidermal keratinocytes constitutively express B7-1 antigen. J Clin Invest. 1994;94:892–8. doi: 10.1172/JCI117411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Williams IR, Ort RJ, Kupper TS. Keratinocyte expression of B7-1 in transgenic mice amplifies the primary immune response to cutaneous antigens. Proc Natl Acad Sci U S A. 1994;91:12780–4. doi: 10.1073/pnas.91.26.12780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ferguson TA, Dube P, Griffith TS. Regulation of contact hypersensitivity by interleukin 10. J Exp Med. 1994;179:1597–604. doi: 10.1084/jem.179.5.1597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Forster R, et al. CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell. 1999;99:23–33. doi: 10.1016/s0092-8674(00)80059-8. [DOI] [PubMed] [Google Scholar]
  • 60.Itano AA, et al. Distinct dendritic cell populations sequentially present antigen to CD4 T cells and stimulate different aspects of cell-mediated immunity. Immunity. 2003;19:47–57. doi: 10.1016/s1074-7613(03)00175-4. [DOI] [PubMed] [Google Scholar]
  • 61.Allenspach EJ, Lemos MP, Porrett PM, Turka LA, Laufer TM. Migratory and lymphoid-resident dendritic cells cooperate to efficiently prime naive CD4 T cells. Immunity. 2008;29:795–806. doi: 10.1016/j.immuni.2008.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Ohl L, et al. CCR7 governs skin dendritic cell migration under inflammatory and steady-state conditions. Immunity. 2004;21:279–88. doi: 10.1016/j.immuni.2004.06.014. [DOI] [PubMed] [Google Scholar]
  • 63.Chorro L, et al. Langerhans cell (LC) proliferation mediates neonatal development, homeostasis, and inflammation-associated expansion of the epidermal LC network. J Exp Med. 2009;206:3089–100. doi: 10.1084/jem.20091586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Merad M, et al. Langerhans cells renew in the skin throughout life under steady-state conditions. Nat Immunol. 2002;3:1135–41. doi: 10.1038/ni852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Romani N, Clausen BE, Stoitzner P. Langerhans cells and more: langerin-expressing dendritic cell subsets in the skin. Immunol Rev. 2010;234:120–41. doi: 10.1111/j.0105-2896.2009.00886.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Henri S, et al. The dendritic cell populations of mouse lymph nodes. J Immunol. 2001;167:741–8. doi: 10.4049/jimmunol.167.2.741. [DOI] [PubMed] [Google Scholar]
  • 67.Bursch LS, et al. Identification of a novel population of Langerin+ dendritic cells. J Exp Med. 2007;204:3147–56. doi: 10.1084/jem.20071966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Ginhoux F, et al. Blood-derived dermal langerin+ dendritic cells survey the skin in the steady state. J Exp Med. 2007;204:3133–46. doi: 10.1084/jem.20071733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Poulin LF, et al. The dermis contains langerin+ dendritic cells that develop and function independently of epidermal Langerhans cells. J Exp Med. 2007;204:3119–31. doi: 10.1084/jem.20071724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Shklovskaya E, Roediger B, Fazekas de St Groth B. Epidermal and dermal dendritic cells display differential activation and migratory behavior while sharing the ability to stimulate CD4+ T cell proliferation in vivo. J Immunol. 2008;181:418–30. doi: 10.4049/jimmunol.181.1.418. [DOI] [PubMed] [Google Scholar]
  • 71.Ginhoux F, et al. The origin and development of nonlymphoid tissue CD103+ DCs. J Exp Med. 2009;206:3115–30. doi: 10.1084/jem.20091756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Henri S, et al. CD207+ CD103+ dermal dendritic cells cross-present keratinocyte-derived antigens irrespective of the presence of Langerhans cells. J Exp Med. 2010;207:189–206. doi: 10.1084/jem.20091964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Edelson BT, et al. Peripheral CD103+ dendritic cells form a unified subset developmentally related to CD8alpha+ conventional dendritic cells. J Exp Med. 2010;207:823–36. doi: 10.1084/jem.20091627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Merad M, Ginhoux F, Collin M. Origin, homeostasis and function of Langerhans cells and other langerin-expressing dendritic cells. Nat Rev Immunol. 2008;8:935–47. doi: 10.1038/nri2455. [DOI] [PubMed] [Google Scholar]
  • 75.Le Borgne M, et al. Dendritic cells rapidly recruited into epithelial tissues via CCR6/CCL20 are responsible for CD8+ T cell crosspriming in vivo. Immunity. 2006;24:191–201. doi: 10.1016/j.immuni.2006.01.005. [DOI] [PubMed] [Google Scholar]
  • 76.Ginhoux F, et al. Langerhans cells arise from monocytes in vivo. Nat Immunol. 2006;7:265–73. doi: 10.1038/ni1307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Kaplan DH, Jenison MC, Saeland S, Shlomchik WD, Shlomchik MJ. Epidermal langerhans cell-deficient mice develop enhanced contact hypersensitivity. Immunity. 2005;23:611–20. doi: 10.1016/j.immuni.2005.10.008. [DOI] [PubMed] [Google Scholar]
  • 78.Bobr A, et al. Acute ablation of Langerhans cells enhances skin immune responses. J Immunol. 2010;185:4724–8. doi: 10.4049/jimmunol.1001802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Igyarto BZ, et al. Langerhans cells suppress contact hypersensitivity responses via cognate CD4 interaction and langerhans cell-derived IL-10. J Immunol. 2009;183:5085–93. doi: 10.4049/jimmunol.0901884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Yoshiki R, et al. IL-10-producing Langerhans cells and regulatory T cells are responsible for depressed contact hypersensitivity in grafted skin. J Invest Dermatol. 2009;129:705–13. doi: 10.1038/jid.2008.304. [DOI] [PubMed] [Google Scholar]
  • 81.Obhrai JS, et al. Langerhans cells are not required for efficient skin graft rejection. J Invest Dermatol. 2008;128:1950–5. doi: 10.1038/jid.2008.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Kautz-Neu K, et al. Langerhans cells are negative regulators of the anti-Leishmania response. J Exp Med. 2011;208:885–91. doi: 10.1084/jem.20102318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Schwarz A, et al. Langerhans cells are required for UVR-induced immunosuppression. J Invest Dermatol. 2010;130:1419–27. doi: 10.1038/jid.2009.429. [DOI] [PubMed] [Google Scholar]
  • 84.Fukunaga A, et al. Langerhans cells serve as immunoregulatory cells by activating NKT cells. J Immunol. 2010;185:4633–40. doi: 10.4049/jimmunol.1000246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kissenpfennig A, et al. Dynamics and Function of Langerhans Cells In Vivo Dermal Dendritic Cells Colonize Lymph Node AreasDistinct from Slower Migrating Langerhans Cells. Immunity. 2005;22:643–654. doi: 10.1016/j.immuni.2005.04.004. [DOI] [PubMed] [Google Scholar]
  • 86.Bennett CL, et al. Inducible ablation of mouse Langerhans cells diminishes but fails to abrogate contact hypersensitivity. J Cell Biol. 2005;169:569–76. doi: 10.1083/jcb.200501071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Bennett CL, Noordegraaf M, Martina CA, Clausen BE. Langerhans cells are required for efficient presentation of topically applied hapten to T cells. J Immunol. 2007;179:6830–5. doi: 10.4049/jimmunol.179.10.6830. [DOI] [PubMed] [Google Scholar]
  • 88.Wang L, et al. Langerin Expressing Cells Promote Skin Immune Responses under Defined Conditions. J Immunol. 2008;180:4722–7. doi: 10.4049/jimmunol.180.7.4722. [DOI] [PubMed] [Google Scholar]
  • 89.Kumamoto Y, Denda-Nagai K, Aida S, Higashi N, Irimura T. MGL2 Dermal dendritic cells are sufficient to initiate contact hypersensitivity in vivo. PLoS One. 2009;4:e5619. doi: 10.1371/journal.pone.0005619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Honda T, et al. Compensatory role of Langerhans cells and langerin-positive dermal dendritic cells in the sensitization phase of murine contact hypersensitivity. J Allergy Clin Immunol. 2010;125:1154–1156 e2. doi: 10.1016/j.jaci.2009.12.005. [DOI] [PubMed] [Google Scholar]
  • 91.Noordegraaf M, Flacher V, Stoitzner P, Clausen BE. Functional redundancy of Langerhans cells and Langerin+ dermal dendritic cells in contact hypersensitivity. J Invest Dermatol. 2010;130:2752–9. doi: 10.1038/jid.2010.223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Bedoui S, et al. Cross-presentation of viral and self antigens by skin-derived CD103+ dendritic cells. Nat Immunol. 2009;10:488–95. doi: 10.1038/ni.1724. [DOI] [PubMed] [Google Scholar]
  • 93.Igyarto BZ, et al. Skin-Resident Murine Dendritic Cell Subsets Promote Distinct and Opposing Antigen-Specific T Helper Cell Responses. Immunity. 2011 doi: 10.1016/j.immuni.2011.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Marshall JS, King CA, McCurdy JD. Mast cell cytokine and chemokine responses to bacterial and viral infection. Curr Pharm Des. 2003;9:11–24. doi: 10.2174/1381612033392413. [DOI] [PubMed] [Google Scholar]
  • 95.Abraham SN, St John AL. Mast cell-orchestrated immunity to pathogens. Nat Rev Immunol. 2010;10:440–52. doi: 10.1038/nri2782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Askenase PW, et al. Defective elicitation of delayed-type hypersensitivity in W/Wv and SI/SId mast cell-deficient mice. J Immunol. 1983;131:2687–94. [PubMed] [Google Scholar]
  • 97.Galli SJ, Hammel I. Unequivocal delayed hypersensitivity in mast cell-deficient and beige mice. Science. 1984;226:710–3. doi: 10.1126/science.6494907. [DOI] [PubMed] [Google Scholar]
  • 98.Tsuji RF, et al. B cell-dependent T cell responses: IgM antibodies are required to elicit contact sensitivity. J Exp Med. 2002;196:1277–90. doi: 10.1084/jem.20020649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Jawdat DM, Albert EJ, Rowden G, Haidl ID, Marshall JS. IgE-mediated mast cell activation induces Langerhans cell migration in vivo. J Immunol. 2004;173:5275–82. doi: 10.4049/jimmunol.173.8.5275. [DOI] [PubMed] [Google Scholar]
  • 100.McLachlan JB, Catron DM, Moon JJ, Jenkins MK. Dendritic cell antigen presentation drives simultaneous cytokine production by effector and regulatory T cells in inflamed skin. Immunity. 2009;30:277–88. doi: 10.1016/j.immuni.2008.11.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Kalesnikoff J, Galli SJ. New developments in mast cell biology. Nat Immunol. 2008;9:1215–23. doi: 10.1038/ni.f.216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Suto H, et al. Mast cell-associated TNF promotes dendritic cell migration. J Immunol. 2006;176:4102–12. doi: 10.4049/jimmunol.176.7.4102. [DOI] [PubMed] [Google Scholar]
  • 103.Bryce PJ, et al. Immune sensitization in the skin is enhanced by antigen-independent effects of IgE. Immunity. 2004;20:381–92. doi: 10.1016/s1074-7613(04)00080-9. [DOI] [PubMed] [Google Scholar]
  • 104.Grimbaldeston MA, Nakae S, Kalesnikoff J, Tsai M, Galli SJ. Mast cell-derived interleukin 10 limits skin pathology in contact dermatitis and chronic irradiation with ultraviolet B. Nat Immunol. 2007;8:1095–104. doi: 10.1038/ni1503. [DOI] [PubMed] [Google Scholar]
  • 105.Galli SJ, Grimbaldeston M, Tsai M. Immunomodulatory mast cells: negative, as well as positive, regulators of immunity. Nat Rev Immunol. 2008;8:478–86. doi: 10.1038/nri2327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Scholten J, et al. Mast cell-specific Cre/loxP-mediated recombination in vivo. Transgenic Res. 2008;17:307–15. doi: 10.1007/s11248-007-9153-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Dudeck A, et al. Mast Cells Are Key Promoters of Contact Allergy that Mediate the Adjuvant Effects of Haptens. Immunity. 2011;34:973–84. doi: 10.1016/j.immuni.2011.03.028. [DOI] [PubMed] [Google Scholar]
  • 108.Strid J, et al. Acute upregulation of an NKG2D ligand promotes rapid reorganization of a local immune compartment with pleiotropic effects on carcinogenesis. Nat Immunol. 2008;9:146–54. doi: 10.1038/ni1556. [DOI] [PubMed] [Google Scholar]
  • 109.Macleod AS, Havran WL. Functions of skin-resident gammadelta T cells. Cell Mol Life Sci. 2011;68:2399–408. doi: 10.1007/s00018-011-0702-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Girardi M, et al. Resident skin-specific gammadelta T cells provide local, nonredundant regulation of cutaneous inflammation. J Exp Med. 2002;195:855–67. doi: 10.1084/jem.20012000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Lewis JM, et al. Selection of the cutaneous intraepithelial gammadelta+ T cell repertoire by a thymic stromal determinant. Nat Immunol. 2006;7:843–50. doi: 10.1038/ni1363. [DOI] [PubMed] [Google Scholar]
  • 112.Toulon A, et al. A role for human skin-resident T cells in wound healing. J Exp Med. 2009;206:743–50. doi: 10.1084/jem.20081787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.DeGroot A. Test Concentrations and vehicles for 2800 allergens. Elsevier; New York, NY: 1986. Patch Testing. [Google Scholar]
  • 114.Vocanson M, Hennino A, Rozieres A, Poyet G, Nicolas JF. Effector and regulatory mechanisms in allergic contact dermatitis. Allergy. 2009;64:1699–714. doi: 10.1111/j.1398-9995.2009.02082.x. [DOI] [PubMed] [Google Scholar]
  • 115.Kupper TS, Fuhlbrigge RC. Immune surveillance in the skin: mechanisms and clinical consequences. Nat Rev Immunol. 2004;4:211–22. doi: 10.1038/nri1310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Akiba H, et al. Skin inflammation during contact hypersensitivity is mediated by early recruitment of CD8+ T cytotoxic 1 cells inducing keratinocyte apoptosis. J Immunol. 2002;168:3079–87. doi: 10.4049/jimmunol.168.6.3079. [DOI] [PubMed] [Google Scholar]
  • 117.Cavani A, et al. Human CD25+ regulatory T cells maintain immune tolerance to nickel in healthy, nonallergic individuals. J Immunol. 2003;171:5760–8. doi: 10.4049/jimmunol.171.11.5760. [DOI] [PubMed] [Google Scholar]
  • 118.Vocanson M, et al. Inducible costimulator (ICOS) is a marker for highly suppressive antigen-specific T cells sharing features of TH17/TH1 and regulatory T cells. J Allergy Clin Immunol. 2010;126:280–9. 289 e1–7. doi: 10.1016/j.jaci.2010.05.022. [DOI] [PubMed] [Google Scholar]
  • 119.O’Leary JG, Goodarzi M, Drayton DL, von Andrian UH. T cell- and B cell-independent adaptive immunity mediated by natural killer cells. Nat Immunol. 2006;7:507–16. doi: 10.1038/ni1332. [DOI] [PubMed] [Google Scholar]
  • 120.von Bubnoff D, et al. Natural killer cells in atopic and autoimmune diseases of the skin. J Allergy Clin Immunol. 2010;125:60–8. doi: 10.1016/j.jaci.2009.11.020. [DOI] [PubMed] [Google Scholar]
  • 121.Carbone T, et al. CD56highCD16-CD62L-NK cells accumulate in allergic contact dermatitis and contribute to the expression of allergic responses. J Immunol. 2010;184:1102–10. doi: 10.4049/jimmunol.0902518. [DOI] [PubMed] [Google Scholar]
  • 122.Balato A, Unutmaz D, Gaspari AA. Natural killer T cells: an unconventional T-cell subset with diverse effector and regulatory functions. J Invest Dermatol. 2009;129:1628–42. doi: 10.1038/jid.2009.30. [DOI] [PubMed] [Google Scholar]
  • 123.Nieuwenhuis EE, et al. CD1d and CD1d-restricted iNKT-cells play a pivotal role in contact hypersensitivity. Exp Dermatol. 2005;14:250–8. doi: 10.1111/j.0906-6705.2005.00289.x. [DOI] [PubMed] [Google Scholar]
  • 124.Campos RA, et al. Cutaneous immunization rapidly activates liver invariant Valpha14 NKT cells stimulating B-1 B cells to initiate T cell recruitment for elicitation of contact sensitivity. J Exp Med. 2003;198:1785–96. doi: 10.1084/jem.20021562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Yusuf N, et al. Protective role of Toll-like receptor 4 during the initiation stage of cutaneous chemical carcinogenesis. Cancer Res. 2008;68:615–22. doi: 10.1158/0008-5472.CAN-07-5219. [DOI] [PMC free article] [PubMed] [Google Scholar]

Hightlighted References

  • 9.Landsteiner K, Jacobs J. Studies on the Sensitization of Animals with Simple Chemical Compounds : Iii. Anaphylaxis Induced by Arsphenamine. J Exp Med. 1936;64:717–21. doi: 10.1084/jem.64.5.717. Comment: This article first proposed the idea that the hapten-self complex is a critical early event in ACD. Seventy-five years later, this hypothesis has been supported by the work of many investigators, and remains a core concept of ACD.
  • 20.Martin SF, et al. Toll-like receptor and IL-12 signaling control susceptibility to contact hypersensitivity. J Exp Med. 2008;205:2151–62. doi: 10.1084/jem.20070509. Comment: This is the first article to suggest that innate immune receptors, in the form of TLR, play an important role in CHS
  • 29. Weber FC, et al. Lack of the purinergic receptor P2X(7) results in resistance to contact hypersensitivity. J Exp Med. 2010;207:2609–19. doi: 10.1084/jem.20092489. Comment: This study demonstrated that self-molecules (ATP) released as a result of cellular damage by haptens activates the inflammasome.
  • 32. Schmidt M, et al. Crucial role for human Toll-like receptor 4 in the development of contact allergy to nickel. Nat Immunol. 2010;11:814–9. doi: 10.1038/ni.1919. Comment: This study demonstrated how the world’s most common contact allergen, Nickel, activates TLR-4, resulting in human APC activation upon exposure to this ubiquitious chemical.
  • 60.Itano AA, et al. Distinct dendritic cell populations sequentially present antigen to CD4 T cells and stimulate different aspects of cell-mediated immunity. Immunity. 2003;19:47–57. doi: 10.1016/s1074-7613(03)00175-4. Comment: This study demonstrated that a productive immune response requires the transport antigen to the region lymph node by skin-resident dendritic cells.
  • 107.Dudeck A, et al. Mast Cells Are Key Promoters of Contact Allergy that Mediate the Adjuvant Effects of Haptens. Immunity. 2011;34:973–84. doi: 10.1016/j.immuni.2011.03.028. Comment: This paper demonstrated that the adjuvant effects of haptens requires mast cells and histamine. Mast cell are also required for hapten-induced DC migration to regional lymph nodes and for CHS

RESOURCES