HHS Public Access Author manuscript Author Manuscript

J Invest Dermatol. Author manuscript; available in PMC 2017 August 10. Published in final edited form as: J Invest Dermatol. 2017 February ; 137(2): 288–294. doi:10.1016/j.jid.2016.08.013.

Emerging Skin T-Cell Functions in Response to Environmental Insults Jutamas Suwanpradid1, Zachary E. Holcomb1,2, and Amanda S. MacLeod1,3,4 1Department 2Duke

of Dermatology, Duke University Medical Center, Durham, North Carolina, USA

University School of Medicine, Durham, North Carolina, USA

Author Manuscript

3Department

of Immunology, Duke University Medical Center, Durham, North Carolina, USA

4Pinnell

Center for Investigative Dermatology and Skin Disease Research Center, Duke University Medical Center, Durham, North Carolina, USA

Abstract Skin is the primary barrier between the body and the outside world, functioning not only as a physical barrier, but also as an immunologic first line of defense. A large number of T cells populate the skin. This review highlights the ability of these cutaneous T cells to regulate skinspecific environmental threats, including microbes, injuries, solar UV radiation, and allergens. Since much of this knowledge has been advanced from murine studies, we focus our review on how the mouse state has informed the human state, emphasizing the key parallels and differences.

Author Manuscript

INTRODUCTION Mouse models have been instrumental in furthering our understanding of cutaneous T-cell biology and function. Importantly, despite the differences of T-cell types and TCR usage between murine and human skin T cells, many of their roles are shared between these two species. The heterogeneity of cutaneous-resident T cells in murine and human skin is highlighted in Table 1.

T CELLS IN MURINE SKIN

Author Manuscript

In a steady state, murine T cells reside predominantly at the epidermal-dermal junction (Figure 1). The epidermis contains a unique population of γδ T cells, called dendritic epidermal T cells (DETCs) (Havran et al., 1989; Tamaki et al., 2001). DETCs reside within the skin near the interface with the skin and the external environment, indicating their role in barrier function and immunity (Girardi et al., 2002, 2006; Jameson et al., 2002; MacLeod et al., 2013; Sharp et al., 2005). DETCs develop in the embryonic thymus and express an invariant VΓ3Vδ1TCR, yet the antigen of the DETC remains elusive. In addition, DETCs

Correspondence: Amanda S. (Büchau) MacLeod, Laboratory for Cutaneous Immunobiology, Department of Dermatology, Duke University Medical Center, Purple Zone, DUMC 3135, 40 Duke Medicine Circle, Durham, North Carolina 27710, USA. [email protected]. CONFLICT OF INTEREST The authors state no conflict of interest.

Suwanpradid et al.

Page 2

Author Manuscript Author Manuscript

also express natural killer group receptor 2D (NKG2D) and can receive signals from NKG2D ligands, expressed by stressed keratinocytes (Nielsen et al., 2015; Strid et al., 2008). In a steady state, DETCs form polarized immunologic synapses that anchor to keratinocyte tight junctions and likely provide TCR-mediated signals for maintenance in the skin (Chodaczek et al., 2012). Despite detailed experimental analyses, a DETC population has not been identified in human skin. The dermis of healthy mice contains both Γδ and αβ T cells. Murine dermal Γδ T cells are self-renewing and are largely maintained independently of circulating precursors (Sumaria et al., 2011). Similar to DETCs, dermal Γδ T cells display an activated memory-like CD44+CD69+CD103+ phenotype;however, they express variant Γδ TCR chains, with some preference for VΓ4 (Sumaria et al., 2011). DETCs require TCR (co-)stimulation, dermal Γδ T cells are highly “innate” and sensitive to cytokine and pathogen-associated molecular pattern recognition (MacLeod et al., 2013; Nielsen et al., 2014, 2015; Strid et al., 2008; Witherden et al., 2010, 2012). Whereas DETCs are dependent on both IL-7 and IL-15 for survival and maintenance, Vγ4+ dermal γδ T cells are largely dependent on IL-7 but not IL-15. A small subset of VΓ4+ dermal Γδ T cells is IL-7 independent (Sumaria et al., 2011; Ye et al., 2001).

Author Manuscript

Under homeostatic conditions, αβ TCR+ T cells comprise approximately 40–50% of all dermal T cells (Cai et al., 2011; Sumaria et al., 2011). Both CD8+ and CD4+ T cells, the latter comprising regulatory T cells (TREG) and nonregulatory T cells, exhibit strong tropism for the hair follicle region (Figure 1), suggesting that the hair follicle is critical in regulating skin residence of these tissue-resident memory T cells (TRM) (Chow et al., 2013; Collins et al., 2016; Gratz et al., 2013). Studies demonstrate that hair follicle epithelial cells produce IL-7 and IL-15, the former cytokine required for both CD4+ and CD8+ TRM persistence, whereas the latter is required for CD8+ TRM epidermotropism (Adachi et al., 2015; Gratz et al., 2013; Lawson et al., 2015). Epidermal TRM are comprised predominantly of CD8+ T cells, and similar to DETCs, these cells typically bear the α-chain of the integrin αEβ7 and CD103, and express the activation marker CD69 (Adachi et al., 2015; Mackay et al., 2013). CD103-mediated retention of T cells in the skin likely occurs via adhesion to E-cadherin. Integrins expressed by the interfollicular and isthmus keratinocytes of the hair follicle activate latent transforming growth factor-β production in the skin, which enhances CD103 expression (Adachi et al., 2015; Casey et al., 2012; El-Asady et al., 2005; Mackay et al., 2013; Masopust et al., 2010; Mohammed et al., 2016).

Author Manuscript

The murine skin also contains CD103−CD8+ TRM cells (Mackay et al., 2013), but the underlying mechanisms of their maintenance in the skin are not well understood. CD69 suppresses sphingosine-1-phosphate receptor 1, a molecule known to prevent T cells from emigrating from lymphoid organs or other tissues into the skin (Skon et al., 2013). Furthermore, CCR4 and CD103 expressed by dermal TREG cells are critical to TREG migratory behavior and maintenance in the skin, respectively (Chow et al., 2013; Suffia et al., 2005). Disruption of the skin barrier induces αβ T-cell migration to and from the lymph node, including effector TEFF, regulatory, and central memory T cells (TCM) (Bromley et al., 2013; Mackay et al., 2013; Tomura et al., 2010), and many of them become long-lived antigen-specific skin-resident memory T cells, termed TRM. The critical roles of αβ and γδ T cells in the contexts of defense against skin infections, host-microbiome interactions, and skin damage will be discussed in this review article. J Invest Dermatol. Author manuscript; available in PMC 2017 August 10.

Suwanpradid et al.

Page 3

Author Manuscript

THE CUTANEOUS T-CELL REPERTOIRE IN HUMAN SKIN

Author Manuscript

Both epidermal and dermal γδ and αβ T cells are present in noninflamed human skin;however, cutaneous γδ T cells are approximately 300 times less abundant than αβ T cells (Clark et al., 2006; Streilein, 1983; Toulon et al., 2009; Watanabe et al., 2015). Cutaneous γδ T cells express predominantly the Vδ1 TCR chain and the skin-homing cutaneous lymphocyte antigen (CLA) (Toulon et al., 2009). TRM participate in immunosurveillance because of their robust and long-lasting memory responses and derive from a surviving effector T cells (TEFF) pool. Human skin TRM comprise CD103+ and CD103− subsets;CD103+ T cells are enriched in the epidermis, whereas CD103− TRM are more frequent in the dermis (Watanabe et al., 2015). Recent studies demonstrate that in addition to TRM, there are two subsets of CCR7+ TCM that can also migrate to the skin. One subset coexpresses L-selectin (CD62L), whereas the other subset does not;the latter subset has been termed migratory memory T cells (Park and Kupper, 2015). In addition, healthy human skin also harbors Foxp3+ memory regulatory T cells, termed mTREG cells, residing around hair follicles where they contribute to immune homeostasis, or if dysregulated can mediate immunopathology (Chow et al., 2013; Sanchez Rodriguez et al., 2014; Seneschal et al., 2012). The composition and function of cutaneous T cells can change dramatically on environmental insults and may be driven by both antigen exposure and innate immune signals derived from surrounding keratinocytes, dendritic cells, macrophages, and other cells (Iwasaki and Medzhitov, 2015; Schroder et al., 2006). This review article focuses on the effects of cutaneous T-cell activation in the context of environmental insults and the factors that maintain T cells in the skin to provide long-lived protective functions.

CONTROL OF COMMENSAL AND PATHOGENIC MICROBIOTA BY SKIN T Author Manuscript

CELLS

Author Manuscript

The cutaneous microbiome is highly diverse and consists of trillions of bacteria, fungi, viruses, archaea, and small arthropods (Byrd and Segre, 2015; Costello et al., 2009; Grice et al., 2009; Human Microbiome Project Consortium, 2012; Kong and Segre, 2012). The composition of the mouse and human skin microbiome differs extensively in both species, but the microbiome exists without generating a host immunologic response. This concept of host-microbiome tolerance in the skin is supported by a recent study in mice showing that pathogen-specific Foxp3-expressing TREG populate the skin postnatally, at a time where commensal colonization of the skin occurs (Scharschmidt et al., 2015). In contrast, dysbiosis is associated with multiple skin diseases, including atopic eczema, and is associated with dysregulated T-cell function (Kobayashi et al., 2015). In humans, commensal microorganisms inhabit the stratum corneum and hair follicle, and possibly even the deeper dermis (Nakatsuji et al., 2013). Changes in the microbiome occur both physiologically and in the setting of skin disease (Kong et al., 2012; Zeeuwen et al., 2012). Based on recent studies, on one hand, the microbiome can enhance protective immunity against pathogens may be driven by cutaneous T cells (and may include additional immune cells). On the other hand, microbiota fine-tune cutaneous T-cell immunity (Naik et al., 2012). Whether the cutaneous microbiome is regulated by antimicrobial peptides and proteins that are inducible by skin T-cell-derived cytokines, including IL-22 and IL-17A, is of particular interest

J Invest Dermatol. Author manuscript; available in PMC 2017 August 10.

Suwanpradid et al.

Page 4

Author Manuscript

(Eyerich et al., 2009; MacLeod et al., 2013; Naik et al., 2012; Sonnenberg et al., 2011). Furthermore, given the recent discovery of a lipid-reactive and CD1a-restricted skin-homing T-cell population producing high levels of IL-22 (de Jong et al., 2014), future studies to determine whether skin lipids derived from microbiota can directly regulate T-cell homing and maintenance in human skin are needed. Elucidating in detail the functional interactions between microbiome diversity and skin T cells in a steady state and in several skin diseases will provide novel strategies for therapies.

Author Manuscript

Multiple mouse models have advanced our understanding of cutaneous protective TRM functions in the context of viral infections, including cutaneous herpes simplex virus (HSV)-1 and vaccinia virus infections. Here, TRM cells reside in the skin during the effector phase of the immune response and persist for several months to provide optimal protection against reinfection by the generation of high levels of IFN-γ. CD8+ TRM primarily accumulate in the epidermis, whereas CD4+ TRM primarily accumulate within the upper dermis (Collins et al., 2016; Gaide et al., 2015; Gebhardt et al., 2009, 2011; Heath and Carbone, 2013; Jiang et al., 2012; Mackay et al., 2012). Keratinocytes are the target cells of most cutaneous viruses; thus, epidermal CD8+ TRM may provide superior protection against viral infections (Mackay et al., 2012; Wakim et al., 2008; Zaid et al., 2014). Epidermal CD8+ TRM residence involves a concomitant local reduction in DETC numbers in the epidermis, indicating that these populations persist in mutual exclusion and may compete for local survival signals (Zaid et al., 2014). Upon viral infection, clusters of IFN-γ-producing CD4+ T cells surrounding hair follicles appear and contain CCL5-producing (APCS), as well CD8+ T cells that increase CD4+ T-cell recruitment into the skin (Collins et al., 2016).

Author Manuscript

In contrast to viral infections, immune surveillance against cutaneous bacterial and fungal infections is largely provided by γδ T cells. Here, cutaneous IL-17-producing γδ T cells are critical to combat murine cutaneous Staphylococcus aureus and Candida albicans infections (Cho et al., 2010; Kashem et al., 2015). However, whether previous infections and induction of IL-17-producing T cells lead to generation of long-term “γδ–TRM” and at least partial protection against reinfection is currently unknown.

Author Manuscript

The functional roles of TRM in human skin in the context of pathogenic infections have been enhanced by ex vivo and in vitro studies, but they are largely correlative in nature. Human skin from newborns, but not from adults, harbors effector T cells that produce high levels of IL-8 (CXCL8) to activate antimicrobial neutrophils and γδ T cells (Gibbons et al., 2014). Adult CLA+ skin-tropic T cells proliferate strongly in vitro after stimulation with pathogens commonly encountered at the cutaneous epithelial surface, such as S. epidermidis, the herpes simplex virus (HSV)-1, and Candida. Notably, in response to C. albicans stimulation, skin-homing T cells isolated from human blood produce IL-9 to enhance effector function of IL-9-producing T helper type 9 (Th9) cells and other effector T cells. Moreover, these Th9 cells reside in healthy human skin and comprise a small but distinct TRM subset (Schlapbach et al., 2014). Furthermore, staphylococcal exotoxins trigger IL-22 production from skinhoming Th22, indicating their potential role in host defense and triggering skin inflammation (Kobayashi et al., 2015; Niebuhr et al., 2010).

J Invest Dermatol. Author manuscript; available in PMC 2017 August 10.

Suwanpradid et al.

Page 5

Author Manuscript

SKIN T CELLS PROMOTE WOUND HEALING AND RE-ESTABLISH THE ANTIMICROBIAL BARRIER

Author Manuscript

Upon skin injury, inflammatory and antimicrobial immune responses are aimed at rapidly clearing microbial contamination before an anti-inflammatory repair program can facilitate wound closure. In mice, γδ T cells, in particular DETCs, have been shown to play critical roles in skin wound repair through recognizing a stress-induced DETC antigen on damaged keratinocytes (Komori et al., 2012), and subsequently stimulating the re-establishment of the skin antimicrobial and physical barrier (Jameson et al., 2002; MacLeod et al., 2013). Whether the previously suggested DETC antigen, Skint-1, truly mediates the stress-induced DETC activation upon wounding needs additional proof, as Skint-1 has not been successfully visualized in the skin nor in the thymus (Barbee et al., 2011). Once activated, a subset of DETCs rapidly produces IL-17A upon skin injury, before the influx of additional peripheral T cells into the skin (Baum and Arpey, 2005; MacLeod et al., 2013), and induces epidermal host-defense molecules, including β-defensin 3, S100A8, and RegIIIg (MacLeod et al., 2013). These antimicrobial peptides and proteins function not only by killing microbes, but also by regulating keratinocyte migration, energy metabolism, cytoskeletal dynamics, and keratinocyte proliferation and differentiation in the skin (Lai et al., 2012; MacLeod et al., 2013; Thorey et al., 2001). Additional cytokines and keratinocyte growth factor are produced by skin-resident γδ T cells in mice to stimulate keratinocyte proliferation (Barbee et al., 2011). Cutaneous TREG cells regulate the transition from wound inflammation to the anti-inflammatory repair phase, as these cells promote wound repair through controlling overt inflammation and proinflammatory macrophage accumulation in murine skin wounds (Nosbaum et al., 2016).

Author Manuscript

Human cutaneous αβ and γδ T cells play pivotal roles in wound repair as well. An elegant study by Toulon et al. (2009) demonstrated that αβ and γδ TCR+ skin-resident T cells isolated from acute, but not from nonhealing chronic wounds, produce insulin-like growth factor 1 upon activation to promote wound repair. Furthermore, human cutaneous T helper type 22 cells produce IL-22, IL-17, and other cytokines. They can stimulate keratinocyte production of antimicrobial peptides and proteins and directly induce keratinocyte proliferation, and collagen and fibronectin production (Eyerich et al., 2009; Lai et al., 2012; McGee et al., 2013). Whether TREG cells also control cutaneous wound repair in humans has not been fully elucidated.

SKIN T CELLS AND UV RADIATION Author Manuscript

Solar UV radiation is a major environmental stressor causing skin damage. The major events studied in the skin after UV exposure are induction of DNA damage, inflammation, immunosuppression, and carcinogenesis. The role of skin T cells in UV-irradiated skin is thus very complex and will be summarized here. UV radiation induces DNA damage through creation of photoproducts and reactive oxygen species, either through direct induction of chemical reactions within DNA or through DNAstrand breaks (Barnes et al., 2010; Gehrke et al., 2013; Greinert et al., 2012; Kripke et al., 1992; Lankinen et al., 1996). UVB radiation (290–320 nm wavelength) and wavelengths

J Invest Dermatol. Author manuscript; available in PMC 2017 August 10.

Suwanpradid et al.

Page 6

Author Manuscript Author Manuscript

close to the UVB spectrum are particularly associated with DNA damage and carry a high risk of inducing genetic instability and cancer (Rünger, 2013). In vivo mouse studies focusing on responses to acute inflammatory UV-induced DNA damage have identified critical functions of DETCs in epidermal DNA damage repair (MacLeod et al., 2014). Mechanistically, UV-irradiated damaged keratinocytes release ATP, which is sensed by purinergic receptors on DETCs and other skin cells, leading to increased DETC IL-17A production. IL-17A induces an epidermal tumor necrosis factor-related weak inducer of apoptosis and growth arrest, and DNA damage-associated gene 45, these two molecules are linked to the DNA repair response and regulation of cell survival and cell death (Hildesheim et al., 2002; MacLeod et al., 2014; Sabour Alaoui et al., 2012). Although this study suggests that DETCs can reduce keratinocyte DNA damage, by limiting γH2AX and cyclobutane pyrimidine dimers, early on, a recent study by Girardi’s group reported that once UVinduced p53 mutations in keratinocytes are acquired, T cells may not be critical in controlling Langerhans cell-driven expansion of these p53 mutant clones (Lewis et al., 2015). However, given the protective cytotoxic role of effector and memory T cells in cancer immunosurveillance (Girardi et al., 2001; Nasti et al., 2011; Strid et al., 2008) and the induction of TREG cells by UV-associated with immunosuppression (Schwarz et al., 2011), it remains to be determined which roles αβ versus γδ T cells play at distinct stages of skin cancer initiation, promotion, and progression.

Author Manuscript

UV radiation has dramatic tolerogenic and immunosuppressive effects on T cells, both locally and systemically. Elegant studies performed in mice have shown that UV-induced tolerance involves the suppression of cutaneous CD4+ and CD8+ effector functions, and this increases TREG activity in the context of allergic contact hypersensitivity and transplantable tumor models (Rana et al., 2008; Schwarz et al., 2005, 2011; Toichi et al., 2002). Together, these studies indicate that maintaining genome integrity in the setting of UV-induced DNA damage, as well as additional immune-protective cytotoxic T-cell functions, is vital for the prevention of early tumor initiation and these protective responses are likely suppressed during skin carcinogenesis (Girardi et al., 2001; MacLeod et al., 2014).

Author Manuscript

Human cutaneous T-cell responses to acute and chronic UV radiation show some parallels to those observed in mice. Both tumor necrosis factor-related weak inducer of apoptosis and growth arrest and DNA damage-associated gene 45 playa role in human skin homeostasis (Maeda et al., 2003; Sabour Alaoui et al., 2012) and are induced by T-cell-derived IL-17A (MacLeod et al., 2014). IL-17A generation is stimulated from human cutaneous TRM by extracellular ATP, a danger signal released upon UV damage (MacLeod et al., 2014). However, future studies will need to clarify in detail the role of human T cells in keratinocyte DNA damage response and how these processes are suppressed in UV-induced skin cancers, such as squamous cell carcinomas and basal cell carcinomas. Similar to mice, UV exposure sufficiently suppresses T-cell-mediated elicitation reactions of contact hypersensitivity responses in human skin (Cooper et al., 1992; Kelly et al., 1998), indicating that there are strong tolerogenic effects of UV on cutaneous TRM. These effects may directly contribute to the development of human squamous cell carcinomas, which are characterized by numeric and qualitative changes of TRM in the skin with increased frequency of TREG cells (Clark et al., 2008; McCray et al., 2015). Because TRM differ between humans and mice (see Table 1), determining which subsets of human cutaneous T cells distinctly J Invest Dermatol. Author manuscript; available in PMC 2017 August 10.

Suwanpradid et al.

Page 7

Author Manuscript

contribute to skin cancer suppression and promotion, and identification of the molecular mechanisms will allow novel targeted therapies.

PATHOLOGIC ROLES FOR SKIN T CELLS IN HYPERSENSITIVITY REACTIONS TO CONTACT ALLERGEN AND DRUGS

Author Manuscript Author Manuscript

Environmental contact allergens such as nickel, perfumes, latex, and poison ivy, can result in T-cell-driven skin inflammation, commonly known as allergic contact dermatitis. Murine delayed contact hypersensitivity (CHS) is an experimental model of human allergic contact dermatitis and consists of two separate phases: sensitization and elicitation (He et al., 2009; Heo et al., 2015; Wang et al., 2000; Xu et al., 1996). The initial sensitization phase is characterized by the formation of complexes between allergens and epidermal proteins, which are subsequently taken up and presented by APCs. These APCs migrate to the draining lymph nodes to present these complexes to naïve T cells, leading them to differentiate into TCM and TRM. These TCM and TRM cells have overlapping TCR repertoires (Gaide et al., 2015) and serve as the major effector cells for mediating the robust inflammatory damaging response in the elicitation phase of CHS. Notably, despite identical TCRs, TRM have a stronger and more rapid response than TCM after re-exposure to the same contact allergen (Gaide et al., 2015). Whether this is simply the result of distinct anatomic locations is currently unknown. In addition to αβ T cells, γδ T cells also play important roles in the murine CHS response. Upon contact allergen exposure, keratinocyte-derived IL-1β triggers IL-17A production in DETCs to mediate inflammation (Nielsen et al., 2014). In contrast, TREG cells serve an immunosuppressive function and negatively regulate the CHS responses via the suppression of T-cell effector functions (Christensen et al., 2015; Gomez de Aguero et al., 2012; Honda et al., 2010). Whether langerin-expressing Langerhans cells, the enigmatic dendritic cell population within the epidermis, or langerin+ dermal dendritic cells activate TREG and play suppressive roles in CHS, or are relevant as APCs at the sensitization phase, remains a matter of debate. Differences and paradoxical findings, however, may be due to differences in allergen applications and/or the timing of ablation of langerin+ cells via diphtheria toxin in the CHS model using langerin-diphtheria toxin receptor (DTR) mice (Bennet et al., 2005, 2007; Gomez de Aguero et al., 2012; Honda et al., 2010; Kaplan et al., 2005; Kissenpfennig et al., 2005).

Author Manuscript

In human skin, CD4+ Th1 and Th2 subsets, along with CD8+ T cells, serve as effector cells in allergic contact dermatitis (Cavani et al., 2001);however, the role of γδ T cells in allergic contact dermatitis in humans is still poorly understood. Notably, cutaneous T cells mediate damage to epidermal keratinocytes, a phenomenon also observed in fixed drug eruptions (FDEs). FDEs are skin lesions occurring after repeated ingestion of causative drugs, and similar to a contact allergy, FDE resembles a cytotoxic reaction of cutaneous CD8+ T cells against epidermal keratinocytes in human skin. Of note, new FDE lesions often develop at the site of a previous viral infection, suggesting that protective antiviral long-lived CD8+ TRM cells in the skin may be reactivated by antigen cross-presentation to subsequently induce FDE lesions. However, FDE cannot be considered a true delayed type IV allergy response such as allergic contact dermatitis, and is instead characterized by the “conversion”

J Invest Dermatol. Author manuscript; available in PMC 2017 August 10.

Suwanpradid et al.

Page 8

Author Manuscript

of lesional CD8+CD45RO+ TRM into CD8+CD45Ra+ T cells in clinically resolved skin (Shiohara et al., 2015).

CONCLUSIONS

Author Manuscript

Functional studies in mice and humans have highlighted the relevance of T cells in cutaneous immunology and maintenance of the body’s first line of defense. Cutaneous memory T cells play pivotal effector roles as “gate keepers” against environmental insults through regulation and protection against microbes, injury, and cancer development. TREG are critical in maintaining immune tolerance against self-antigens and promoting skin repair. Dysregulation and overly vigorous activation of skin T cells can cause detrimental effects, resulting in skin pathology. The heterogeneity of skin T cells is just beginning to be understood, and future studies are required to further define skin T-cell subsets and their anatomical, structural, and functional relationships. Novel therapeutic strategies to either inhibit or augment skin T-cell function are being developed for clinical use, and specific targeting of these cells holds promise for the treatment of a variety of human skin conditions, including autoimmune diseases, infections, and cancer.

Acknowledgments We apologize to all authors whose work could not be cited here due to space limitations. ASM is supported by the NIH through award K08AR063729 05. As a Duke Cancer Institute member, we gratefully acknowledge grantsupport from the Duke Cancer Institute (to ASM) as part of the P30 Cancer Center Support Grant (P30CA014236). As a member of the Skin Disease Research Center and Pinnell Center for Investigative Dermatology, we gratefully acknowledge grant support (to ASM) from the Duke Skin Disease Research Center (P30AR066527 02) and Pinnell Center for Investigative Dermatology and Skin Disease Research Center at Duke University Medical Center.

Author Manuscript

Abbreviations

Author Manuscript

APC

antigen-presenting cell

CHS

contact hypersensitivity

CLA

cutaneous lymphocyte-associated antigen

DETC

dendritic epidermal T cell

FDE

fixed drug eruption

TCM

central memory T cell

TEFF

effector T cell

Th

T helper

TREG

regulatory T cell

TRM

tissue-resident memory T cell

J Invest Dermatol. Author manuscript; available in PMC 2017 August 10.

Suwanpradid et al.

Page 9

Author Manuscript

References

Author Manuscript Author Manuscript Author Manuscript

Adachi T, Kobayashi T, Sugihara E, Yamada T, Ikuta K, Pittaluga S, et al. Hair follicle-derived IL-7 and IL-15 mediate skin-resident memory T cell homeostasis and lymphoma. Nat Med. 2015; 21:1272–9. [PubMed: 26479922] Barbee SD, Woodward MJ, Turchinovich G, Mention JJ, Lewis JM, Boyden LM, et al. Skint-1 is a highly specific, unique selecting component for epidermal T cells. Proc Natl Acad Sci USA. 2011; 108:3330–5. [PubMed: 21300860] Barnes L, Dumas M, Juan M, Noblesse E, Tesniere A, Schnebert S, et al. γH2AX, an accurate marker that analyzes UV genotoxic effects on human keratinocytes and on human skin. Photochem Photobiol. 2010; 86:933–41. [PubMed: 20492564] Baum CL, Arpey CJ. Normal cutaneous wound healing: clinical correlation with cellular and molecular events. Dermatol Surg. 2005; 31:674–86. discussion 86. [PubMed: 15996419] Bennet CL, Noordegraaf M, Martina CA, Clausen BE. Langerhans cells are required for efficient presentation to topically applied hapten to T cells. J Immunol. 2007; 179:6830–5. [PubMed: 17982073] Bennet CL, van Rijn E, Inaba K, Steinman RM, Kapsenberg ML, Clausen B. Inducible ablation of mouse Langerhans cells fails to abrogate contact hypersensitivity. J Cell Biol. 2005; 169:569–76. [PubMed: 15897263] Bromley SK, Yan S, Tomura M, Kanagawa O, Luster AD. Recirculating memory T cells are a unique subset of CD4+ T cells with a distinct phenotype and migratory pattern. J Immunol. 2013; 190:970– 6. [PubMed: 23255361] Byrd AL, Segre JA. Integrating host gene expression and the microbiome to explore disease pathogenesis. Genome Biol. 2015; 16:70. [PubMed: 25887564] Cai Y, Shen X, Ding C, Qi C, Li K, Li X, et al. Pivotal role of dermal IL-17-producing γδ T cells in skin inflammation. Immunity. 2011; 35:596–610. [PubMed: 21982596] Casey KA, Fraser KA, Schenkel JM, Moran A, Abt MC, Beura LK, et al. Antigen-independent differentiation and maintenance of effector-like resident memory T cells in tissues. J Immunol. 2012; 188:4866–75. [PubMed: 22504644] Cavani A, Albanesi C, Traidl C, Sebastiani S, Girolomoni G. Effector and regulatory T cells in allergic contact dermatitis. Trends Immunol. 2001; 22:118–20. [PubMed: 11286716] Cho JS, Pietras EM, Garcia NC, Ramos RI, Farzam DM, Monroe HR, et al. IL-17 is essential for host defense against cutaneous Staphylococcus aureus infection in mice. J Clin Invest. 2010; 120:1762– 73. [PubMed: 20364087] Chodaczek G, Papanna V, Zal MA, Zal T. Body-barrier surveillance by epidermal γδ TCRs. Nat Immunol. 2012; 13:272–82. [PubMed: 22327568] Chow Z, Mueller SN, Deane JA, Hickey MJ. Dermal regulatory T cells display distinct migratory behavior that is modulated during adaptive and innate inflammation. J Immunol. 2013; 191:3049– 56. [PubMed: 23940277] Christensen AD, Skov S, Kvist PH, Haase C. Depletion of regulatory T cells in a hapten-induced inflammation model results in prolonged and increased inflammation driven by T cells. Clin Exp Immunol. 2015; 179:485–99. [PubMed: 25302741] Clark RA, Chong B, Mirchandani N, Brinster NK, Yamanaka K, Dowgiert RK, et al. The vast majority of CLA+ T cells are resident in normal skin. J Immunol. 2006; 176:4431–9. [PubMed: 16547281] Clark RA, Huang SJ, Murphy GF, Mollet IG, Hijnen D, Muthukuru M, et al. Human squamous cell carcinomas evade the immune response by down-regulation of vascular E-selectin and recruitment of regulatory T cells. J Exp Med. 2008; 205:2221–34. [PubMed: 18794336] Collins N, Jiang X, Zaid A, Macleod BL, Li J, Park CO, et al. Skin CD4(+) memory T cells exhibit combined cluster-mediated retention and equilibration with the circulation. Nat Commun. 2016; 7:11514. [PubMed: 27160938] Cooper KD, Oberhelman L, Hamilton TA, Baadsgaard O, Terhune M, LeVee G, et al. UV exposure reduces immunization rates and promotes tolerance to epicutaneous antigens in humans: relationship to dose, CD1a-DR+ epidermal macrophage induction, and Langerhans cell depletion. Proc Natl Acad Sci USA. 1992; 89:8497–501. [PubMed: 1382291]

J Invest Dermatol. Author manuscript; available in PMC 2017 August 10.

Suwanpradid et al.

Page 10

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI, Knight R. Bacterial community variation in human body habitats across space and time. Science. 2009; 326:1694–7. [PubMed: 19892944] de Jong A, Cheng TY, Huang S, Gras S, Birkinshaw RW, Kasmar AG, et al. CD1a-autoreactive T cells recognize natural skin oils that function as headless antigens. Nat Immunol. 2014; 15:177–85. [PubMed: 24362891] El-Asady R, Yuan R, Liu K, Wang D, Gress RE, Lucas PJ, et al. TGF-{beta}-dependent CD103 expression by CD8(+) T cells promotes selective destruction of the host intestinal epithelium during graft-versus-host disease. J Exp Med. 2005; 201:1647–57. [PubMed: 15897278] Eyerich S, Eyerich K, Pennino D, Carbone T, Nasorri F, Pallotta S, et al. Th22 cells represent a distinct human T cell subset involved in epidermal immunity and remodeling. J Clin Invest. 2009; 119:3573–85. [PubMed: 19920355] Gaide O, Emerson RO, Jiang X, Gulati N, Nizza S, Desmarais C, et al. Common clonal origin of central and resident memory T cells following skin immunization. Nat Med. 2015; 21:647–53. [PubMed: 25962122] Gebhardt T, Wakim LM, Eidsmo L, Reading PC, Heath WR, Carbone FR. Memory T cells in nonlymphoid tissue that provide enhanced local immunity during infection with herpes simplex virus. Nat Immunol. 2009; 10:524–30. [PubMed: 19305395] Gebhardt T, Whitney PG, Zaid A, Mackay LK, Brooks AG, Heath WR, et al. Different patterns of peripheral migration by memory CD4+ and CD8+ T cells. Nature. 2011; 477:216–9. [PubMed: 21841802] Gehrke N, Mertens C, Zillinger T, Wenzel J, Bald T, Zahn S, et al. Oxidative damage of DNA confers resistance to cytosolic nuclease TREX1 degradation and potentiates STING-dependent immune sensing. Immunity. 2013; 39:482–95. [PubMed: 23993650] Gibbons D, Fleming P, Virasami A, Michel ML, Sebire NJ, Costeloe K, et al. Interleukin-8 (CXCL8) production is a signatory T cell effector function of human newborn infants. Nat Med. 2014; 20:1206–10. [PubMed: 25242415] Girardi M, Lewis JM, Filler RB, Hayday AC, Tigelaar RE. Environmentally responsive and reversible regulation of epidermal barrier function by gammadelta T cells. J Invest Dermatol. 2006; 126:808– 14. [PubMed: 16439970] Girardi M, Lewis J, Glusac E, Filler RB, Geng L, Hayday AC, et al. Resident skin-specific gammadelta T cells provide local, nonredundant regulation of cutaneous inflammation. J Exp Med. 2002; 195:855–67. [PubMed: 11927630] Girardi M, Oppenheim DE, Steele CR, Lewis JM, Glusac E, Filler R, et al. Regulation of cutaneous malignancy by gammadelta T cells. Science. 2001; 294:605–9. [PubMed: 11567106] Gomez de Aguero M, Vocanson M, Hacini-Rachinel F, Taillardet M, Sparwasser T, Kissenpfennig A, et al. Langerhans cells protect from allergic contact dermatitis in mice by tolerizing CD8(+) T cells and activating Foxp3(+) regulatory T cells. J Clin Invest. 2012; 122:1700–11. [PubMed: 22523067] Gratz IK, Truong HA, Yang SH, Maurano MM, Lee K, Abbas AK, et al. Cutting edge: memory regulatory t cells require IL-7 and not IL-2 for their maintenance in peripheral tissues. J Immunol. 2013; 190:4483–7. [PubMed: 23543753] Greinert R, Volkmer B, Henning S, Breitbart E, Greulich K, Cardoso M, et al. UVA-induced DNA double-strand breaks result from the repair of clustered oxidative DNA damages. Nucleic Acids Res. 2012; 40:10263–73. [PubMed: 22941639] Grice EA, Kong HH, Conlan S, Deming CB, Davis J, Young AC, et al. Topographical and temporal diversity of the human skin microbiome. Science. 2009; 324:1190–2. [PubMed: 19478181] Havran WL, Grell S, Duwe G, Kimura J, Wilson A, Kruisbeek AM, et al. Limited diversity of T-cell receptor gamma-chain expression of murine Thy-1+ dendritic epidermal cells revealed by V gamma 3-specific monoclonal antibody. Proc Natl Acad Sci USA. 1989; 86:4185–9. [PubMed: 2726770] He D, Wu L, Kim HK, Li H, Elmets CA, Xu H. IL-17 and IFN-gamma mediate the elicitation of contact hypersensitivity responses by different mechanisms and both are required for optimal responses. J Immunol. 2009; 183:1463–70. [PubMed: 19553527]

J Invest Dermatol. Author manuscript; available in PMC 2017 August 10.

Suwanpradid et al.

Page 11

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Heath WR, Carbone FR. The skin-resident and migratory immune system in steady state and memory: innate lymphocytes, dendritic cells and T cells. Nat Immunol. 2013; 14:978–85. [PubMed: 24048119] Heo WI, Lee KE, Hong JY, Kim MN, Oh MS, Kim YS, et al. The role of interleukin-17 in mouse models of atopic dermatitis and contact dermatitis. Clin Exp Dermatol. 2015; 40:665–71. [PubMed: 25684357] Hildesheim J, Bulavin DV, Anver MR, Alvord WG, Hollander MC, Vardanian L, et al. GADD45a protects against UV irradiation-induced skin tumors, and promotes apoptosis and stress signaling via MAPK and p53. Cancer Res. 2002; 62:7305–15. [PubMed: 12499274] Honda T, Nakajima S, Egawa G, Ogasawara K, Malissen B, Miyachi Y, 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. [PubMed: 20226508] Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature. 2012; 486:207–14. [PubMed: 22699609] Iwasaki A, Medzhitov R. Control of adaptive immunity by the innate immune system. Nat Immunol. 2015; 16:343–53. [PubMed: 25789684] Jameson J, Ugarte K, Chen N, Yachi P, Fuchs E, Boismenu R, et al. A role for skin gammadelta T cells in wound repair. Science. 2002; 296:747–9. [PubMed: 11976459] Jiang X, Clark RA, Liu L, Wagers AJ, Fuhlbrigge RC, Kupper TS. Skin infection generates nonmigratory memory CD8+ T(RM) cells providing global skin immunity. Nature. 2012; 483:227–31. [PubMed: 22388819] Kaplan DH, Jenison MC, Saeland S, Shlomchik WD, Shlomchik MJ. Epidermal Langerhans celldeficient mice develop enhanced contact hypersensitivity. Immunity. 2005; 23:611–20. [PubMed: 16356859] Kashem SW, Igyarto BZ, Gerami-Nejad M, Kumamoto Y, Mohammed J, Jarrett E, et al. Candida albicans morphology and dendritic cell subsets determine T helper cell differentiation. Immunity. 2015; 42:356–66. [PubMed: 25680275] Kelly DA, Walker SL, McGregor JM, Young AR. A single exposure of solar simulated radiation suppresses contact hypersensitivity responses both locally and systemically in humans: quantitative studies with high-frequency ultrasound. J Photochem Photobiol B. 1998; 44:130–42. [PubMed: 9757595] Kissenpfennig A, Henri S, Dubois B, Laplace-Builhe C, Perrin P, Romani N, et al. Dynamics and function of Langerhans cells in vivo: dermal dendritic cells colonize lymph node areas distinct from slower migrating Langerhans cells. Immunity. 2005; 22:643–54. [PubMed: 15894281] Kobayashi T, Glatz M, Horiuchi K, Kawasaki H, Akiyama H, Kaplan DH, et al. Dysbiosis and Staphylococcus aureus colonization drives inflammation in atopic dermatitis. Immunity. 2015; 42:756–66. [PubMed: 25902485] Komori HK, Witherden DA, Kelly R, Sendaydiego K, Jameson JM, Teyton L, et al. Cutting edge: dendritic epidermal gammadelta T cell ligands are rapidly and locally expressed by keratinocytes following cutaneous wounding. J Immunol. 2012; 188:2972–6. [PubMed: 22393149] Kong HH, Oh J, Deming C, Conlan S, Grice EA, Beatson MA, et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012; 22:850–9. [PubMed: 22310478] Kong HH, Segre JA. Skin microbiome: looking back to move forward. J Invest Dermatol. 2012; 132(Pt 2):933–9. [PubMed: 22189793] Kripke ML, Cox PA, Alas LG, Yarosh DB. Pyrimidine dimers in DNA initiate systemic immunosuppression in UV-irradiated mice. Proc Natl Acad Sci USA. 1992; 89:7516–20. [PubMed: 1502162] Lai Y, Li D, Li C, Muehleisen B, Radek KA, Park HJ, et al. The antimicrobial protein REG3A regulates keratinocyte proliferation and differentiation after skin injury. Immunity. 2012; 37:74–84. [PubMed: 22727489] Lankinen MH, Vilpo LM, Vilpo JA. UV-and γ-irradiation-induced DNA single-strand breaks and their repair in human blood granulocytes and lymphocytes. Mutat Res. 1996; 352:31–8. [PubMed: 8676912]

J Invest Dermatol. Author manuscript; available in PMC 2017 August 10.

Suwanpradid et al.

Page 12

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Lawson BR, Gonzalez-Quintial R, Eleftheriadis T, Farrar MA, Miller SD, Sauer K, et al. Interleukin-7 is required for CD4(+) T cell activation and autoimmune neuroinflammation. Clin Immunol. 2015; 161:260–9. [PubMed: 26319414] Lewis JM, Burgler CD, Freudzon M, Golubets K, Gibson JF, Filler RB, et al. Langerhans cells facilitate UVB-induced epidermal carcinogenesis. J Invest Dermatol. 2015; 135:2824–33. [PubMed: 26053049] Mackay LK, Rahimpour A, Ma JZ, Collins N, Stock AT, Hafon ML, et al. The developmental pathway for CD103(+)CD8+ tissue-resident memory T cells of skin. Nat Immunol. 2013; 14:1294–301. [PubMed: 24162776] Mackay LK, Stock AT, Ma JZ, Jones CM, Kent SJ, Mueller SN, et al. Long-lived epithelial immunity by tissue-resident memory T (TRM) cells in the absence of persisting local antigen presentation. Proc Natl Acad Sci USA. 2012; 109:7037–42. [PubMed: 22509047] MacLeod AS, Hemmers S, Garijo O, Chabod M, Mowen K, Witherden DA, et al. Dendritic epidermal T cells regulate skin antimicrobial barrier function. J Clin Invest. 2013; 123:4364–74. [PubMed: 24051381] MacLeod AS, Rudolph R, Corriden R, Ye I, Garijo O, Havran WL. Skin-resident T cells sense ultraviolet radiation-induced injury and contribute to DNA repair. J Immunol. 2014; 192:5695– 702. [PubMed: 24808367] Maeda T, Sim AB, Leedel DA, Chua PP, Chomey EG, Luong L, et al. UV induces GADD45 in a p53dependent and -independent manner in human keratinocytes. J Cutan Med Surg. 2003; 7:119–23. [PubMed: 12447616] Masopust D, Choo D, Vezys V, Wherry EJ, Duraiswamy J, Akondy R, et al. Dynamic T cell migration program provides resident memory within intestinal epithelium. J Exp Med. 2010; 207:553–64. [PubMed: 20156972] McCray C, Cook J, Streilein RD, Degan S, Havran WL, Zhang J, et al. ENTPD1 and PD-1/PD-1 ligands are expressed by skin-resident immune cells in subsets of human SCC. J Invest Dermatol. 2015; 135:S1–8. McGee HM, Schmidt BA, Booth CJ, Yancopoulos GD, Valenzuela DM, Murphy AJ, et al. IL-22 promotes fibroblast-mediated wound repair in the skin. J Invest Dermatol. 2013; 133:1321–9. [PubMed: 23223145] Mohammed J, Beura LK, Bobr A, Astry B, Chicoine B, Kashem SW, et al. Stromal cells control the epithelial residence of DCs and memory T cells by regulated activation of TGF-beta. Nat Immunol. 2016; 17:414–21. [PubMed: 26901152] Naik S, Bouladoux N, Wilhelm C, Molloy MJ, Salcedo R, Kastenmuller W, et al. Compartmentalized control of skin immunity by resident commensals. Science. 2012; 337:1115–9. [PubMed: 22837383] Nakatsuji T, Chiang HI, Jiang SB, Nagarajan H, Zengler K, Gallo RL. The microbiome extends to subepidermal compartments of normal skin. Nat Commun. 2013; 4:1431. [PubMed: 23385576] Nasti TH, Iqbal O, Tamimi IA, Geise JT, Katiyar SK, Yusuf N. Differential roles of T-cell subsets in regulation of ultraviolet radiation induced cutaneous photocarcinogenesis. Photochem Photobiol. 2011; 87:387–98. [PubMed: 21143237] Niebuhr M, Scharonow H, Gathmann M, Mamerow D, Werfel T. Staphylococcal exotoxins are strong inducers of IL-22: A potential role in atopic dermatitis. J Allergy Clin Immunol. 2010; 126:1176– 1183.e4. [PubMed: 20864149] Nielsen MM, Dyring-Andersen B, Schmidt JD, Witherden D, Lovato P, Woetmann A, et al. NKG2Ddependent activation of dendritic epidermal T cells in contact hypersensitivity. J Invest Dermatol. 2015; 135:1311–9. [PubMed: 25634359] Nielsen MM, Lovato P, MacLeod AS, Witherden DA, Skov L, DyringAndersen B, et al. IL-1betadependent activation of dendritic epidermal T cells in contact hypersensitivity. J Immunol. 2014; 192:2975–83. [PubMed: 24600030] Nosbaum A, Prevel N, Truong HA, Mehta P, Ettinger M, Scharschmidt TC, et al. Cutting edge: regulatory T cells facilitate cutaneous wound healing. J Immunol. 2016; 196:2010–4. [PubMed: 26826250]

J Invest Dermatol. Author manuscript; available in PMC 2017 August 10.

Suwanpradid et al.

Page 13

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Park CO, Kupper TS. The emerging role of resident memory T cells in protective immunity and inflammatory disease. Nat Med. 2015; 21:688–97. [PubMed: 26121195] Rana S, Byrne SN, MacDonald LJ, Chan CY-Y, Halliday GM. Ultraviolet B suppresses immunity by inhibiting effector and memory T cells. Am J Pathol. 2008; 172:993–1004. [PubMed: 18292235] Rünger TM. Much remains to be learned about how UVR induces mutations. J Invest Dermatol. 2013; 133:1717–9. [PubMed: 23760050] Sabour Alaoui S, Dessirier V, de Araujo E, Alexaki VI, Pelekanou V, Lkhider M, et al. TWEAK affects keratinocyte G2/M growth arrest and induces apoptosis through the translocation of the AIF protein to the nucleus. PLoS One. 2012; 7:e33609. [PubMed: 22438963] Sanchez Rodriguez R, Pauli ML, Neuhaus IM, Yu SS, Arron ST, Harris HW, et al. Memory regulatory T cells reside in human skin. J Clin Invest. 2014; 124:1027–36. [PubMed: 24509084] Scharschmidt TC, Vasquez KS, Truong HA, Gearty SV, Pauli ML, Nosbaum A, et al. A wave of regulatory T cells into neonatal skin mediates tolerance to commensal microbes. Immunity. 2015; 43:1011–21. [PubMed: 26588783] Schlapbach C, Gehad A, Yang C, Watanabe R, Guenova E, Teague JE, et al. Human Th9 cells are skintropic and have autocrine and paracrine proinflammatory capacity. Sci Transl Med. 2014; 6:219ra8. Schroder JM, Reich K, Kabashima K, Liu FT, Romani N, Metz M, et al. Who is really in control of skin immunity under physiological circumstances—lymphocytes, dendritic cells or keratinocytes? Exp Dermatol. 2006; 15:913–29. [PubMed: 17002689] Schwarz A, Maeda A, Kernebeck K, van Steeg H, Beissert S, Schwarz T. Prevention of UV radiationinduced immunosuppression by IL-12 is dependent on DNA repair. J Exp Med. 2005; 201:173–9. [PubMed: 15657287] Schwarz A, Navid F, Sparwasser T, Clausen BE, Schwarz T. In vivo reprogramming of UV radiation— induced regulatory T-cell migration to inhibit the elicitation of contact hypersensitivity. J Allergy Clin Immunol. 2011; 128:826–33. [PubMed: 21762977] Seneschal J, Clark RA, Gehad A, Baecher-Allan CM, Kupper TS. Human epidermal Langerhans cells maintain immune homeostasis in skin by activating skin resident regulatory T cells. Immunity. 2012; 36:873–84. [PubMed: 22560445] Sharp LL, Jameson JM, Cauvi G, Havran WL. Dendritic epidermal T cells regulate skin homeostasis through local production of insulin-like growth factor 1. Nat Immunol. 2005; 6:73–9. [PubMed: 15592472] Shiohara T, Ushigome Y, Kano Y, Takahashi R. Crucial role of viral reactivation in the development of severe drug eruptions: a comprehensive review. Clin Rev Allergy Immunol. 2015; 49:192–202. [PubMed: 24736996] Skon CN, Lee JY, Anderson KG, Masopust D, Hogquist KA, Jameson SC. Transcriptional downregulation of S1pr1 is required for the establishment of resident memory CD8+ T cells. Nat Immunol. 2013; 14:1285–93. [PubMed: 24162775] Sonnenberg GF, Fouser LA, Artis D. Border patrol: regulation of immunity, inflammation and tissue homeostasis at barrier surfaces by IL-22. Nat Immunol. 2011; 12:383–90. [PubMed: 21502992] Streilein JW. Skin-associated lymphoid tissues (SALT): origins and functions. J Invest Dermatol. 1983; 80(Suppl):12s–6s. [PubMed: 6602189] Strid J, Roberts SJ, Filler RB, Lewis JM, Kwong BY, Schpero W, 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. [PubMed: 18176566] Suffia I, Reckling SK, Salay G, Belkaid Y. A role for CD103 in the retention of CD4+CD25+ Treg and control of Leishmania major infection. J Immunol. 2005; 174:5444–55. [PubMed: 15845457] Sumaria N, Roediger B, Ng LG, Qin J, Pinto R, Cavanagh LL, et al. Cutaneous immunosurveillance by self-renewing dermal gammadelta T cells. J Exp Med. 2011; 208:505–18. [PubMed: 21339323] Tamaki K, Sugaya M, Tada Y, Yasaka N, Uehira M, Nishimoto H, et al. Epidermal and dermal gammadelta T cells. Chem Immunol. 2001; 79:43–51. [PubMed: 11478154] Thorey IS, Roth J, Regenbogen J, Halle JP, Bittner M, Vogl T, et al. The Ca2+-binding proteins S100A8 and S100A9 are encoded by novel injuryregulated genes. J Biol Chem. 2001; 276:35818– 25. [PubMed: 11463791] J Invest Dermatol. Author manuscript; available in PMC 2017 August 10.

Suwanpradid et al.

Page 14

Author Manuscript Author Manuscript Author Manuscript

Toichi E, McCormick TS, Cooper KD. Cell surface and cytokine phenotypes of skin immunocompetent cells involved in ultraviolet-induced immunosuppression. Methods. 2002; 28:104–10. [PubMed: 12231194] Tomura M, Honda T, Tanizaki H, Otsuka A, Egawa G, Tokura Y, et al. Activated regulatory T cells are the major T cell type emigrating from the skin during a cutaneous immune response in mice. J Clin Invest. 2010; 120:883–93. [PubMed: 20179354] Toulon A, Breton L, Taylor KR, Tenenhaus M, Bhavsar D, Lanigan C, et al. A role for human skinresident T cells in wound healing. J Exp Med. 2009; 206:743–50. [PubMed: 19307328] Wakim LM, Waithman J, van Rooijen N, Heath WR, Carbone FR. Dendritic cell-induced memory T cell activation in nonlymphoid tissues. Science. 2008; 319:198–202. [PubMed: 18187654] Wang B, Fujisawa H, Zhuang L, Freed I, Howell BG, Shahid S, et al. CD4+ Th1 and CD8+ type 1 cytotoxic T cells both play a crucial role in the full development of contact hypersensitivity. J Immunol. 2000; 165:6783–90. [PubMed: 11120799] Watanabe R, Gehad A, Yang C, Scott LL, Teague JE, Schlapbach C, et al. Human skin is protected by four functionally and phenotypically discrete populations of resident and recirculating memory T cells. Sci Transl Med. 2015; 7:279ra39. Witherden DA, Verdino P, Rieder SE, Garijo O, Mills RE, Teyton L, et al. The junctional adhesion molecule JAML is a costimulatory receptor for epithelial gammadelta T cell activation. Science. 2010; 329:1205–10. [PubMed: 20813954] Witherden DA, Watanabe M, Garijo O, Rieder SE, Sarkisyan G, Cronin SJ, et al. The CD100 receptor interacts with its plexin B2 ligand to regulate epidermal gammadelta T cell function. Immunity. 2012; 37:314–25. [PubMed: 22902232] Xu H, DiIulio NA, Fairchild RL. T cell populations primed by hapten sensitization in contact sensitivity are distinguished by polarized patterns of cytokine production: interferon gammaproducing (Tc1) effector CD8+ T cells and interleukin (Il) 4/Il-10-producing (Th2) negative regulatory CD4+ T cells. J Exp Med. 1996; 183:1001–12. [PubMed: 8642241] Ye SK, Maki K, Lee HC, Ito A, Kawai K, Suzuki H, et al. Differential roles of cytokine receptors in the development of epidermal gamma delta T cells. J Immunol. 2001; 167:1929–34. [PubMed: 11489972] Zaid A, Mackay LK, Rahimpour A, Braun A, Veldhoen M, Carbone FR, et al. Persistence of skinresident memory T cells within an epidermal niche. Proc Natl Acad Sci USA. 2014; 111:5307– 12. [PubMed: 24706879] Zeeuwen PL, Boekhorst J, van den Bogaard EH, de Koning HD, van de Kerkhof PM, Saulnier DM, et al. Microbiome dynamics of human epidermis following skin barrier disruption. Genome Biol. 2012; 13:R10. [PubMed: 22348269]

Author Manuscript J Invest Dermatol. Author manuscript; available in PMC 2017 August 10.

Suwanpradid et al.

Page 15

Author Manuscript Author Manuscript

Figure 1. Murine and human skin harbors T cells

The illustration of diverse cutaneous T cell populations in the human and mouse. DETC, dendritic epidermal T cell; TRM, tissue-resident memory T cell.

Author Manuscript Author Manuscript J Invest Dermatol. Author manuscript; available in PMC 2017 August 10.

Suwanpradid et al.

Page 16

Table 1

Author Manuscript

Heterogeneity of human and murine skin resident T cells Location

T-cell subset

Mouse

Epidermis

γδ T cells

Epidermis

CD4+ T cell

CD103+ > CD103− TRM

CD103+ > CD103− TRM

Epidermis

CD4+ TREG

FoxP3+ TREG

FoxP3+ TREG

Epidermis

CD8+ T cell

CD103+ > CD103− TRM

CD103+ > CD103− TRM

Dermis

γδ T cells

Vγ4−/+

Vδ1+ enriched

Dermis

CD4+ T cell

FoxP3− TRM

CD103− > CD103+ TRM

Dermis

CD4+ TREG

FoxP3+ mTREG

FoxP3+ TREG

Dermis

CD8+ T cell

CD103− > CD103+ TRM

CD103− > CD103+ TRM

DETC

(Vγ3+Vδ1+)

Human Vδ1+ enriched

Author Manuscript

Abbreviations: DETC, dendritic epidermal T cell; TREG, regulatory T cell; TRM, tissue-resident memory T cell.

Author Manuscript Author Manuscript J Invest Dermatol. Author manuscript; available in PMC 2017 August 10.

Emerging Skin T-Cell Functions in Response to Environmental Insults.

Skin is the primary barrier between the body and the outside world, functioning not only as a physical barrier, but also as an immunologic first line ...
400KB Sizes 0 Downloads 9 Views