Prévia do material em texto
The skin is a barrier organ that separates the body from the outside environment. Because many physical, chemical and microbial insults affect the skin, various types of immune cells reside in or are recruited into the skin to maintain skin homeostasis upon inflammatory challenges. In the epidermis, Langerhans cells (LCs), which are a unique subset of professional antigen-presenting cells (APCs), reside between keratinocytes (Fig. 1a). Although LCs were previously considered a subset of dendritic cells (DCs) because they migrate to the lymph nodes, recent ontogenic studies have revealed that LCs are actua- lly a subset of tissue-resident macrophages that acquire a DC-like phenotype and functions upon further differ- entiation in the skin1,2. In addition to LCs, two specific subsets of T cells can be found in the mouse epidermis: γδ T cells and CD8+ resident memory T (TRM) cells. γδ T cells are a subset of innate immune cells that reside perma- nently in the epidermis of mice but not humans. CD8+ TRM cells comprise populations of non-circulating memory T cells that appear after the resolution of skin inflam- mation, such as that caused by herpes simplex virus (HSV) infection3. TRM cells have been identified in var- ious non-lymphoid tissues4,5, and their longevity differs between tissues. For example, skin TRM cells in mice per- sist for over a year6, whereas lung TRM cells are maintained for only a few months7. Anatomically, the most unique feature of the epider- mis is the existence of the stratum corneum8,9 (Box 1; Fig. 1a). The stratum corneum is the outermost layer of the epidermis and consists of piles of dead keratino- cytes (known as corneocytes) and intercellular lipids. This structure blocks the entry or exit of water and water-soluble substances; thus, it is essential for animals to survive outside of water. Its barrier function is, how- ever, imperfect because it contains many holes for skin appendages, such as hair follicles and sweat ducts. Skin appendages are useful to protect the body from mechan- ical damage, ultraviolet light, temperature changes and dryness. Thus, animals develop skin appendages at the expense of the integral stratum corneum. It is of note that numerous microorganisms (known as the microbiota) live on the surfaces of our body in a com- mensal relationship and may influence our cutaneous immune reactions10,11. As skin appendages lack the stratum corneum, the skin microbiota coexists beside living keratinocytes in these areas11. This results in skin appendages being immunologically unique spots. In the dermis, there are several types of innate immune cells, including dermal DCs, macrophages, mast cells, γδ T cells and innate lymphoid cells (ILCs)12. In the steady state, small numbers of neutrophils, mono- cytes and αβ T cells survey the dermis for pathogens; in response to inflammatory stimuli, many more immune cells rapidly accumulate in the dermis13–15. Anatomically, the dermis is characterized by an abundant extracellu- lar matrix (ECM), comprising collagen and elastin fibres that fill the extracellular spaces. The ECM provides a scaffold for immune cell migration16. Throughout the ECM, mesh-like networks of blood and lymph vascu- latures and neurons are distributed. Immune cells are constantly recruited to the skin via the blood vascular systems. The recruited cells include the precursor cells of skin-resident DCs and macrophages as well as some pas- senger cells, such as neutrophils and T cells, which scout for foreign pathogens. Dermal DCs that have captured The immunological anatomy of the skin Kenji Kabashima 1,2*, Tetsuya Honda1, Florent Ginhoux 2,3 and Gyohei Egawa1* Abstract | The skin is the outermost organ of the body and is continuously exposed to external pathogens. Upon inflammation, various immune cells pass through, reside in or are recruited to the skin to orchestrate diverse cutaneous immune responses. To achieve this, immune cells interact with each other and even communicate with non-immune cells, including peripheral nerves and the microbiota. Immunologically important anatomical sites, such as skin appendages (for example, hair follicles and sweat glands) or postcapillary venules, act as special portal sites for immune cells and for establishing tertiary lymphoid structures, including inducible skin-associated lymphoid tissue. Here, we provide an overview of the key findings and concepts of cutaneous immunity in association with skin anatomy and discuss how cutaneous immune cells fine-tune physiological responses in the skin. 1Department of Dermatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan. 2Singapore Immunology Network (SIgN) and Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore. 3Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China. *e-mail: kaba@ kuhp.kyoto-u.ac.jp; gyohei@ kuhp.kyoto-u.ac.jp https://doi.org/10.1038/ s41577-018-0084-5 NATuRe RevIeWS | IMMuNoLogy R e v i e w s volume 19 | JANuARY 2019 | 19 http://orcid.org/0000-0002-0773-0554 http://orcid.org/0000-0002-2857-7755 mailto:kaba@ kuhp.kyoto-u.ac.jp mailto:kaba@ kuhp.kyoto-u.ac.jp mailto:gyohei@kuhp.kyoto-u.ac.jp mailto:gyohei@kuhp.kyoto-u.ac.jp https://doi.org/10.1038/s41577-018-0084-5 https://doi.org/10.1038/s41577-018-0084-5 FAS ligand Specific ECM components Treg cell and/or CD4+ TRM cell Infundibulum Isthmus IL-17A Neutrophil T H 17 cells T H 17 cells IL-17A Bulge Demodex sp. (mite) Monocyte and/or macrophage CD8+ T RM cell CCL20 and/or IL-15 CCL8 IL-10 TGFβ IL-1β TGFβ IL-6 CCL2 and/or IL-17 Microbiota Lymph duct DC P. acnes T RM cell LC Sweat duct Stratum corneum Epidermis Hair shaft a b c Stratum corneum Stratum granulosum Stratum spinosum Stratum basale Lymph duct Subcutaneous adipose tissue Dermis Blood vessel γδ T cell Keratinocyte Tight junction Sebaceous gland Sweat gland Fig. 1 | Physical and immunological barrier of the skin. a | Structures of the skin. In the epidermis, tight junctions are formed underneath the stratum corneum (in the stratum granulosum), and three types of immune cell populations (Langerhans cells (LCs), γδ T cells and resident memory T (TRM) cells) reside between keratinocytes. b | A schematic of immune privilege of hair follicles. Hair follicles express FAS ligand while lacking MHC class II expression, and they are surrounded by immune-suppressive extracellular matrix (ECM) components. TRM cells and regulatory T (Treg) cells are found in the vicinity of hair follicles. Numerous microorganisms live deep inside the hair follicles. Follicular keratinocytes in different parts of the follicle produce distinct chemokines and control the traffic of leukocytes in the skin. c | A schematic of immune modulation by sebaceous glands. The bacterium Propionibacterium acnes stimulates sebaceous glands to produce IL-1β, IL-6 and transforming growth factor-β (TGFβ), which leads to the activation of dermal dendritic cells (DCs) that can preferentially prime T helper 17 (TH17) cells. The TH17 cells promote neutrophil recruitment and inflammation at the hair follicle. CCL , CC-chemokine ligand. www.nature.com/nri R e v i e w s 20 | JANuARY 2019 | volume 19 cutaneous antigens migrate back to the draining lymph nodes via the lymphatics17. Neurons endow a sensory function to the skin and can directly communicate with immune cells18,19. Recent developments in imaging studies in vivo have demonstrated the dynamic behaviour of immune cells in the skin and underscored the unique distribution pattern of each immune subset that is related to skin anatomical structures. Thus, knowledge of the interaction between immune cells and non-immune structures is important for a deep understanding of the basic mechanisms of cutaneous immune reactions. In this Review, we dis-cuss the unique nature of some anatomical parts and non-immune components of the skin (for example, hair follicles, blood vessels, neurons, sebaceous glands and the microbiota) and focus on the function of a specific immunological unit, called inducible skin-associated lymphoid tissue (iSALT)20, which has an essential role in the induction of cutaneous adaptive immunity. Immunological anatomy of the epidermis The main function of keratinocytes is to maintain the physical barrier of the skin by developing into the stra- tum corneum. This is achieved by keratinocytes passing through three inner cell layers: the stratum basale, the stratum spinosum and the stratum granulosum (Fig. 1). Keratinocytes are not merely a progenitor of the bar- rier but also a component of the innate immune sys- tem. They express several different pattern recognition receptors and produce a variety of cytokines, including thymic stromal lymphopoietin (TSLP), tumour necro- sis factor (TNF), IL-33 and other IL-1 family members, upon inflammation21. These cytokines are essential for activation of skin-resident immune cells and for immune cell recruitment to the skin (reviewed previously22). As skin appendages lack the stratum corneum, they are points of entry for external pathogens. In addition to low-molecular-weight compounds, such as haptens, living microorganisms (for example, bacteria, fungi, viruses, parasites and mites such as Demodex spp.) can easily reach the vicinity of keratinocytes living in these areas11 (Fig. 1b). To handle this unique situation, hair follicles are equipped with specific immunological properties, as described below. Monocyte interaction with hair follicles. Previous in vivo imaging studies demonstrated that hair folli- cles interact closely with recruited monocytes23. When LCs were depleted by diphtheria toxin treatment of Langerin-DTR mice, GR1high monocyte-derived LC pre- cursor cells appeared around hair follicles within weeks23. This suggested that hair follicles are portal sites where LC precursors enter the epidermis. Such LC repopulation is dependent on CC-chemokine receptor 2 (CCR2) and CCR6, suggesting that hair fol- licles constitutively produce chemoattractants for LCs. Fluorescence-activated cell sorting-based cell isolation and gene expression analyses revealed that the chemo- kine expression patterns of follicular keratinocytes differ according to their location. Specifically, keratinocytes in the hair follicle infundibulum produce CC-chemokine ligand 20 (CCL20); keratinocytes in the isthmus produce CCL2; and keratinocytes around the hair bulge region produce CCL8 (Fig. 1b). Monocyte-derived cells also rally around hair fol- licles during inflammation. These cells have several confusing synonyms and have been referred to as inflammatory monocytes, inflammatory macrophages and inflammatory DCs. When entering the epider- mis, they are called inflammatory dendritic epidermal cells24. After the induction of skin inflammation by hap- ten painting, CX3C-chemokine receptor 1 (CX3CR1)+ monocyte-derived cells form clusters around hair fol- licles within a day in a CCR2-dependent manner25. Although the immunological function of these clus- ters remains unclear, the activation of T cells and their production of IFNγ is facilitated by cluster formation, suggesting that antigen presentation in the skin under inflammatory conditions depends, at least in part, on activated inflammatory monocytes. T cell interaction with hair follicles. In contrast to the folliculotropic properties of monocytes, hair follicles have a repellent nature towards effector T cells in the steady state, which has led the hair follicle to be classified as a site of immune privilege26 (Fig. 1b). This is charac- terized by the downregulation of MHC class I expres- sion in follicular keratinocytes, the local production of potent immunosuppressants (such as transforming growth factor-β1 (TGFβ1) and IL-10), the establish- ment of a unique ECM surrounding the hair follicle and the production of FAS ligands to delete autoreac- tive FAS-expressing T cells. Why hair follicles have this immune privilege is still under discussion, but it likely leads to the sequestration of not only self-antigens in hair follicles but also colonized microorganisms, pre- venting their exposure to T cells. As such, the collapse of immune privilege at these sites and the development of T cell responses against the skin microbiota may potentially result in autoimmune responses against hair follicles, such as those observed in alopecia areata27. By contrast to their repellent effect on effector T cells, hair follicles harbour skin-resident TRM cells, which are responsible for long-term skin immunity28. CD4+ and CD8+ TRM cells predominantly reside around hair fol- licles. Keratinocytes in the infundibulum and isthmus produce IL-15, which is required for the persistence of Langerin-DTR mice Mice that express diphtheria toxin receptor (DTR) under the control of the Langerin gene promoter. Treatment of these mice with diphtheria toxin leads to the deletion of all Langerin-expressing cells. Alopecia areata A patchy hair loss mainly occurring in the scalp. it is believed to be one of the autoimmune diseases. Box 1 | Stratum corneum The stratum corneum is the outermost layer of the epidermis, and it has a central role in maintaining the barrier function of the skin. Its thickness ranges from 10 to 30 μm and varies substantially between body sites. In the stratum corneum, keratinocytes become flattened and denucleated and are called corneocytes; their membranes are replaced by a unique barrier structure known as the cornified envelope8,88. Intercellular spaces are filled with lipids. These structures are often described as the ‘bricks’ (corneocytes) and ‘mortar’ (intercellular lipids), which together provide a highly hydrophobic barrier against the environment. Importantly, the stratum corneum is not formed at skin appendages, which thus provides an outside-to-inside route for small-molecule drugs and chemicals and an inside-to-outside route for transepidermal water loss. NATuRe RevIeWS | IMMuNoLogy R e v i e w s volume 19 | JANuARY 2019 | 21 CD8+ TRM cells in the epidermis. By contrast, IL-7, which is required for both CD4+ and CD8+ TRM cells, is pre- dominantly produced by infundibulum keratinocytes. Another study has shown that CD11b+ cells (most likely macrophages and/or DCs) and CD8+ T cells around the hair follicles produce CCL5, which is proposed to recruit memory CD4+ T cells to form clusters29. During second- ary immune responses, most IFNγ-producing T cells localize around the hair follicles, suggesting that these clusters may protect against pathogens by efficiently acti- vating effector T cells. Because hair follicles facilitate the trafficking of DCs23 as well as T cells28, they may serve as an essential site for antigen presentation in the skin under certain conditions. As for CD4+ regulatory T (Treg) cells, they are pre- dominantly distributed near the hair bulge area where hair follicular stem cells reside. Intriguingly, the deple- tion of skin-resident Treg cells results in the perturbation of hair cycles30. These findings suggest that Treg cells can control hair cycles by interacting with hair follicu- lar stem cells and that they are essential for preserving immune privilege in the follicles. Immune modulation by sebaceous glands. Sebaceous glands are another unique structure that accompanies hair follicles (Fig. 1c). These glands secrete lipids as part of the skin barrier function and produce antimicrobial pep- tides and cytokines and chemokines that modulate skin immunity. In pathological conditions, such as acne, IL-17- producing T helper 17 (TH17) cells accumulate around sebaceous glands31. In vitro analysis demonstrated that Propionibacterium acnes induced sebocytes to produce IL-6, TGFβ and IL-1β, which activateddermal DCs to preferentially prime TH17 cells31. In another pathologi- cal context — eosinophilic pustular folliculitis — sebaceous glands were surrounded by a massive number of eosin- ophils32. In this condition, prostaglandin D2 (PGD2) expression is upregulated in hair follicles, which acts on sebocytes to produce the eosinophil chemoattractant CCL26. It is evident that there are many potential ways in which sebocytes can modulate skin immune responses. Influence of the skin microbiota. Numerous microor- ganisms cover skin surfaces and reside in skin append- ages (Fig. 1b,c). These microorganisms have a profound effect on immune processes of the skin11. The metabo- lites and/or structural components of microorganisms may influence both innate and adaptive immunity. For example, Staphylococcus epidermidis, which constitutes a major population of commensal bacteria found on the skin, produces lipoteichoic acid that acts selectively on keratinocytes through Toll-like receptor 3 (TLR3) to inhibit inflammatory cytokine release33. S. epidermidis is also directly captured by dermal APCs, probably at skin appendages, and antigens from the bacteria are pre- sented on MHC class I molecules by APCs in the drain- ing lymph nodes34. As a result, S. epidermidis-specific CD8+ T cells that express IL-17A or IFNγ (and have been referred to as Tc17 or Tc1 cells, respectively) are recruited to the skin in the absence of overt inflammation34. It is of note that S. epidermidis-specific CD8+ T cells have a unique gene expression profile in that they express genes associated with immunoregulation, angiogenesis, tissue remodelling and ECM production. Consistent with this, S. epidermidis-specific CD8+ T cells promote rapid keratinocyte proliferation and accelerate wound healing. As corneocytes are dead cells and express no active biological sensors for microorganisms, microorgan- isms in the skin appendages are likely to have a greater influence on cutaneous immunity than microorganisms residing on the stratum corneum. Indeed, the compo- sition of the microbiota varies substantially between the surface areas and the deeper areas of the skin35. For the same reason, pathogenic microorganisms target skin appendages unless barrier-disrupted injury exists. Thus, it should be emphasized that the method used to sample skin microbiota, such as swab, tape stripping and skin biopsy, is important to consider when interpreting data from skin microbiota studies35. The expansion of pathogenic bacteria and fungi in skin appendages easily stimulates neighbouring keratinocytes to produce inflammatory cytokines and chemokines that can recruit neutrophils and monocytes, leading to the development of folliculitis and hidrade- nitis. By contrast, pathogenic viruses that invade skin appendages sometimes establish latent infection and form their ‘nest’ in these sites (for example, human pap- illoma viruses), probably owing to the repellent nature of these appendages against T cells36. How the cutaneous immune system discriminates pathogenic microorgan- isms from commensal microorganisms has yet to be fully elucidated, and characterizing the critical molecules required for this recognition would be of clinical interest. Immune responses in the dermis Immune modulation by dermal blood vessels. Dermal blood vessels can be divided into four different parts with distinct functions: arteries, capillaries, postcapil- lary venules and collecting venules (Fig. 2). Among these vessels, postcapillary venules have a unique property that is particularly important during inflammation. Adjacent blood endothelial cells are sealed with tight junctions and adherence junctions. These barrier structures limit the passage of plasma proteins larger than 70 kDa, sug- gesting that extravasation of albumin and immunoglob- ulins (which have molecular masses of approximately 70 kDa and 150 kDa, respectively) is limited under homeostatic conditions37 (Fig. 2). Importantly, this vascu- lar permeability is variable only at postcapillary venules. During inflammation, intercellular tight and/or adher- ence junctions are diminished, and immunoglobulins, albumin and water shift into the dermal interstitium and lead to tissue swelling. This phenomenon demonstrates that postcapillary venules are specific portal sites that allow key mediators of humoral immunity to access the skin under inflammatory conditions (Fig. 2). In postcapillary venules, endothelial cells are sur- rounded by the basement membrane and pericytes, and macrophages and mast cells are located nearby. Recent studies using a mouse model of Staphylococcus aureus infection have demonstrated that these cellular and matrix components act as integrated functional units, termed the ‘perivascular extravasation units’38 (Fig. 2). In these units, perivascular macrophages are Eosinophilic pustular folliculitis A recurrent folliculitis that is often formed in the face. in this condition, many eosinophils are pathologically accumulated around hair follicles. www.nature.com/nri R e v i e w s 22 | JANuARY 2019 | volume 19 critically important for neutrophil extravasation. Perivascular macrophages express high levels of the neutrophil-attracting chemokines CXC-chemokine ligand 1 (CXCL1) and CXCL2, and neutrophils extra- vasate in the vicinity of perivascular macrophages. These macrophages are depleted when they are exposed to the S. aureus-derived exotoxin α-haemolysin, and in the absence of perivascular macrophages, neutrophil recruitment to the skin is markedly suppressed38. Postcapillary venules also play a crucial role dur- ing T cell recruitment to and activation in the skin20 (Fig. 2). During cutaneous inflammation, T cells, DCs and perivascular macrophages form clusters, which have been termed iSALT, around postcapillary venules. The iSALT provides a site of antigen presentation in the skin, which is critical for the elicitation of adaptive immunity. The induction and function of iSALT will be discussed in detail in a later section. Neuro-immune interactions in the skin. The skin func- tions as both a barrier and a sensory interface. It is inner- vated by abundant sensory nerves and also by a smaller number of autonomic nerve fibres. Neurons form mesh-like bundles in the dermis, and a large number of the nerve ends reach the epidermis (Fig. 3a). Whole-mount immune staining analysis revealed that a large fraction of dermal DCs are in close contact with neurons, suggest- ing there is neural regulation of DC functions19. Until recently, it was unclear whether peripheral neurons reg- ulate cutaneous immune responses. However, studies involving the genetic and pharmacological ablation of transient receptor potential subfamily V member 1 (TRPV1)- expressing sensory neurons have demonstrated that neurons have key roles in regulating cutaneous immune responses19,39. For example, skin inflammation induced by the TLR7 ligand imiquimod is significantly suppressed with the pharmacological ablation of TRPV1+ sensory nerves19. Topical application of imiquimod causes a skin inflam- mation that is similar to human psoriasis, particularly in terms of the dependence of the response on the IL-17– IL-23 axis40. Following nerve deletion, dermal DCs fail to produce IL-23 in imiquimod-exposed skin and, con- sequently, the local production of IL-17 by dermal γδ T cells is significantly suppressed40. In a Candida albicans infection model, TRPV1+ sensory nociceptors directly sense fungal agents and release calcitonin gene-related peptide (CGRP); this peptide upregulates the production of IL-23 by CD301b+ DCs, which in turn drives IL-17A production by dermal γδ T cells18 (Fig. 3a). In contrast to these pro-inflammatory roles for cuta- neous neurons, immunoregulatory roles for neurons have been reported in the squaric acid dibutylester (SADBE)-induced model of contact hypersensitivity (CHS)41 (Box 2). Upon SADBEapplication to the skin, TRPV1-expressing neurons directly recognize this hapten and suppress the pro-inflammatory function of dermal macrophages. By contrast, when TRPV1-deficient mice are treated with SADBE, dermal macrophages abundantly produce pro-inflammatory cytokines, such as TNF, IL-1β and IL-6, and this markedly exacerbates skin inflamma- tion (Fig. 3a). These findings suggest that neurons are important immune regulators that can either promote or suppress skin inflammation depending on the context. Immunity in subcutaneous adipose tissue Below the dermis, subcutaneous adipose tissue is distrib- uted throughout the body. It acts as padding, an energy reserve and insulation for thermoregulation42. Adipose tissue contains a variety of immune cells, including T cells, B cells and macrophages. In the obese condi- tion, however, the number of macrophages is increased by fourfold compared with the lean condition43, and the production of a variety of inflammatory media- tors, termed adipokines, is increased. These mediators include adiponectin, leptin, IL-6 and TNF. Subcutaneous adipose tissue is also essential for host defence against Immunoglobulin Low-molecular-weight proteins (70 kDa) Stable state b Inflammatory state Water Water Arteriole Capillaries Postcapillary venules Collecting venules a T cell DC Immunoglobulins Perivascular macrophage Neutrophils A unit for neutrophil extravasation Hyper-permeability for humoral immunity iSALT for T cell activation Fig. 2 | A specific contribution of postcapillary venules in cutaneous immunity. a | Skin blood vessels can be divided into four distinct types: arterioles, capillaries, postcapillary venules and collecting venules. Among them, postcapillary venules display unique properties during inflammation. Inducible skin-associated lymphoid tissues (iSALT) are induced for the activation of T cells that have infiltrated the skin. Perivascular extravasation units are essential for neutrophil recruitment. Perivascular macrophages are critically important for neutrophil extravasation. Hyper-permeabilization occurs in postcapillary venules, and this enables the recruitment of immunoglobulins from the blood. b | A schematic of hyper-permeabilization of postcapillary venules is shown. In the steady state, only plasma components with a molecular mass of 70 kDa), such as albumin and immunoglobulins, freely pass through the hyper-permeabilized blood vessel walls. DC, dendritic cell. Transient receptor potential subfamily V member 1 (TRPV1). Also known as capsaicin receptor; TRPV1 is a cation channel member selectively expressed on peripheral sensory neurons that serves as a molecular sensor (nociceptor) for noxious stimuli. NATuRe RevIeWS | IMMuNoLogy R e v i e w s volume 19 | JANuARY 2019 | 23 S. aureus infection via the production of cathelicidin, which kills bacteria44. This suggests diverse roles for adipocytes in cutaneous immunity. Recent in vivo imaging analysis revealed that adi- pocytes form clusters around dermal blood vessels, which are termed perivascular adipose tissues (PATs)45 (Fig. 3b). The functions of PATs in the skin remain unre- solved, but they may be involved in vascular regula- tion or could provide a harbour for specific subsets of immune cells, analogous to the role played by PATs in other organs46,47. Skin anatomy and induction of adaptive immunity Before eliciting adaptive immune responses in the skin, external antigens breach the skin barrier and are captured by cutaneous DCs. Anatomically, the barrier function of the skin is dependent on two key structures, the stratum corneum and the tight junction8,9 (Fig. 1). As described above, skin appendages lack the stratum corneum, and the tight junction is thus the primary bar- rier in these tissues. As skin appendages act as ‘shunt routes’ into the body for drugs and chemicals, the tight junction is considered the determinant of skin barrier functions. Most contact dermatitis is caused by haptens. Haptens are small molecules that acquire antigenicity only when binding to self-proteins in the skin (lead- ing to the formation of hapten–self complexes). The tight junction limits the passage of molecules larger than 500 Da, and the molecular weight of haptens is less than this. As such, haptens can easily pass through the tight junction and penetrate the skin under phys- iological conditions (Fig. 4). By contrast, macromole- cules, such as proteins, are too large to penetrate tight junctions and cannot diffuse into the underlying skin tissues. However, during inflammation, activated LCs elongate their dendrites outward beyond the tight junction and uptake external antigens48. This is another shunt route into the skin, mediated by LCs, that plays a crucial role in percutaneous sensitization by macromolecular proteins. Antigen presentation by cutaneous APCs. The antigens that breach the stratum corneum and tight junctions are then captured by the second line of the immunologi- cal barrier, cutaneous DCs. DCs are a diverse family of cells that play a crucial role in linking our innate and Dermal DC Neuron Dermal γδ T cells Blood vessel Perivascular adipose tissue LC Tight junction a b Macrophages HaptensFungi and/or bacteria IL-17ACGRP IL-23 Stratum corneum Stratum granulosum Stratum spinosum Stratum basale Fig. 3 | Neuro-immune interactions and perivascular adipose tissue in the skin. a | A schematic of neuro-immune interactions in the skin. Components of fungi and bacteria as well as haptens can be directly sensed by transient receptor potential subfamily V member 1 (TRPV1)+ nociceptors. The activated TRPV1+ nerves produce calcitonin gene-related peptide (CGRP) to upregulate the production of IL-23 by CD301b+ dendritic cells (DCs), which in turn drives IL-17A production by dermal γδ T cells. TRPV1+ nerves also recognize haptens and modulate skin inflammation by regulating the function of dermal macrophages. b | Perivascular adipose tissue in the skin. BODIPY (green) and fluorescent-conjugated dextran (red) were simultaneously injected to visualize lipophilic cells and blood vessels, respectively. Second-harmonic generation represents dermal collagen fibres (blue). LC, Langerhans cell. Box 2 | Contact hypersensitivity responses The contact hypersensitivity (CHS) response provides a murine model of allergic contact dermatitis. It is induced by small compounds called haptens. Haptens are conjugated to self-proteins and are captured by cutaneous dendritic cells (DCs). DCs carry the hapten– self complex to the draining lymph nodes and mediate antigen presentation to prime antigen-specific effector T cells, namely, cytotoxic CD8+ T cells and T helper 1 cells (known as the sensitization phase)73,89,90. With subsequent exposure to the same haptens, activation of the effector T cells is induced by antigen-presenting cells in the skin, leading to spongiotic dermatitis (known as the elicitation phase)68. www.nature.com/nri R e v i e w s 24 | JANuARY 2019 | volume 19 adaptive immune system49. Generally, four cell types were classified in the DC family in the skin: epidermal LCs, conventional DCs (cDCs), plasmacytoid DCs and monocyte-derived DCs. cDCs can be further subdi- vided into the cDC1 and cDC2 subsets on the basis of subset-specific gene expression profiles, their depend- ence on different transcription factors and unique subset functions50,51. During inflammation and neoplasm formation in the skin (Box 3), effector T cells are recruited and retained in the skin with limited antigen dependency52. The skin-infiltrating effector T cells actively migrate in order to explore antigens presented by cutaneous APCs53. Once they encounter their cognate antigens, effector T cells initiate stable contact with APCs and are activatedto produce inflammatory cytokines52,54,55. The motility and the activation status of effector T cells are inversely correlated and finely regulated52. In the elicitation of CHS, dermal DCs are the most important APCs in the skin and form clusters for effi- cient antigen presentation to T cells20 (Fig. 5). In addition to dermal DCs, several other cell populations, such as mast cells and LCs, are proposed as possible APCs in the skin. Mast cells participate in antigen presentation in the skin in CHS by acquiring MHC class II molecules from DCs56. Basophils may also act as APCs in the skin because they can also acquire MHC class II molecules by trogocytosis and present antigens to induce TH2 cells in the lymph nodes57. Although the role of LCs as APCs in the elicitation of CHS remains unclear, LCs may have potential APC functions in a graft versus host disease model, where the depletion of LCs in the effector phase causes impaired CD8+ T cell activation in the skin58. In addition, a recent report using mice with transgenic expression of a fragment of the human gene encoding CD1a on LCs demonstrated that LCs can present lipid antigens to effector T cells in the skin and amplify the inflammatory response in allergic contact dermatitis and psoriasis59. These reports suggest the diverse poten- tial of LCs as APCs in the skin. Furthermore, the loss of MHC class I on radioresistant cells led to impaired granzyme B production in CD8+ T cells in the elicitation phase of CHS60, suggesting the possible involvement of endothelial cells61 and keratinocytes62,63 as APCs in the skin, although in vivo evidence for this remains lack- ing. As co-stimulatory molecules are not required for activation of effector T cells64, various cells may carry out APC functions in the skin in a context-dependent manner (TABLe 1). iSALT formation around postcapillary venules. Recently, a leukocyte-clustering structure named iSALT was proposed to serve as a site for efficient anti- gen presentation in the skin20 (Fig. 5). Around 1980, the antigen-presenting capability of the skin was identified and people offered the term skin-associated lymphoid tissue (SALT) to describe this function65–67. Although SALT is a conceptual term, iSALT is an actual structure that transiently appears around postcapillary venules in response to various immunogenic stimuli, including hap- tens and infection68,69. The iSALT is composed of various types of leukocytes, including perivascular macrophages, dermal DCs and T cells20. After hapten application to the skin, dermal DCs exhibit cluster formation around post- capillary venules within hours. IL-1α produced by kerat- inocytes in response to external insults activates M2-like macrophages around postcapillary venules, which then produce CXCL2 and recruit dermal DCs to the cluster. Leukotriene B4 (LTB4), a lipid mediator, also mediates the cluster formation by promoting DC migration70. Subsequently, effector T cells accumulate in the cluster and are presented antigens by dermal DCs, leading to their proliferation and activation. In the cluster, both major dermal DC subsets71, namely, CD11b+ dermal DCs and CD103+ dermal DCs, are detected72. It is cur- rently unclear, however, which dermal DC subsets medi- ate antigen presentation in the cluster60. Each DC subset in the cluster may work in a compensatory manner D er m is Hapten (FITC) Protein antigen FITC–OVA Claudin 1 DAPI Ep id er m is D erm is Epiderm is Tight junction Fig. 4 | Penetration of hapten and proteins into the skin. A hapten (fluorescein isothiocyanate (FITC); molecular mass = 389; left) or FITC-conjugated ovalbumin (FITC– OVA ; molecular mass = 66 kDa; right) were painted onto the ear skin. After 24 hours, the ears were harvested and sliced. Nuclei (blue) and claudin 1 (red) were visualized. FITC distributed into the dermis (left), probably mainly through the hair follicles and cracks in the stratum corneum, while FITC–OVA remained outside the tight junctions (right). DAPI, 4′,6-diamidino-2-phenylindole. Box 3 | Antigen presentation in skin neoplasms Antigen presentation by antigen-presenting cells and the subsequent activation of cytotoxic CD8+ T cells in the tumour site are essential for antitumour immunity. The blockade of programmed cell death 1 (PD1) signalling is believed to exert antitumour effects by inducing CD8+ T cell activation in the tumour52,91. In cancer, tumour-associated macrophages, CD11b+ dermal dendritic cells (DCs) and CD103+ dermal DCs can interact with CD8+ T cells and restrict the motility of the cells92,93. In melanoma, however, it has been reported that only CD103+ DCs can effectively activate the CD8+ T cells in the tumour94,95. As CD103+ dermal DCs constitute only approximately 1% of the total tumour DCs, the trapping of CD8+ T cells by CD11b+ dermal DCs and tumour-associated macrophages would limit the chance for CD8+ T cells to meet CD103+ dermal DCs and be activated, which may lead to inefficient antitumour immune responses. Because tumour-associated macrophages express PD1, which inhibits phagocytosis and the tumour immunity of tumour-associated macrophages96, PD1 inhibitors may exert their antitumour effects by affecting tumour-associated macrophages. Trogocytosis Lymphocytes that conjugate to antigen-presenting cells sometimes ‘rob’ the surface molecules and express them on their own surfaces. ‘Trogo’ means ‘gnaw’ in greek. NATuRe RevIeWS | IMMuNoLogy R e v i e w s volume 19 | JANuARY 2019 | 25 Sensitization Elicitation CD103+ dermal DC IFNγ CD11b+ dermal DC Skin-draining lymph node 12–24 hours Dermal DC Effector T cell a b • Antigen presentation • Effector T cell differentiation (Tc1 and/or T H 1 cell) Stratum corneum Stratum granulosum Stratum spinosum Stratum basale LC Hapten Epidermis Dermis Effector T cell CXCL2LTB4 Perivascular macrophage Postcapillary venule IL-1α Migration • Antigen presentation • Effector T cell activation Tight junction IFNγ Oedema and� vesicle formation Dermal DC • iSALT formation • Effector T cell activation Fig. 5 | A schematic view of the sensitization and elicitation phases of contact hypersensitivity. a | In the sensitization phase of contact hypersensitivity , haptens pass through the skin barrier (stratum corneum and tight junction) and are captured by cutaneous dendritic cells (DCs), including Langerhans cells (LCs) and dermal DCs. DCs, especially CD103+ dermal DCs, present antigens to naive T cells and induce the differentiation of effector T cells — most notably IFNγ-producing cytotoxic T cells (which have been referred to as Tc1 cells) and T helper 1 (TH1) cells — in the skin-draining lymph nodes. In the elicitation phase, haptens captured by dermal DCs are presented to effector T cells in the skin. The activated effector T cells produce IFNγ and elicit skin inflammation. b | Haptens induce IL-1α production from keratinocytes, which activates M2-type macrophages that are located around postcapillary venules. The activated macrophages then produce CXC-chemokine ligand 2 (CXCL2), which promotes the accumulation of dermal DCs. Leukotriene B4 (LTB4) also plays an important role in driving DC accumulation by increasing DC motility. Effector T cells are activated within the DC clusters (present in inducible skin-associated lymphoid tissue (iSALT)) and produce cytokines. www.nature.com/nri R e v i e w s 26 | JANuARY 2019 | volume 19 like DCs in the sensitization phase73. Nevertheless, the blockade of CXCL2 and IL-1 receptor signal- ling impairs the formation of leukocyte clusters and effector T cell activation, indicating that iSALT is an essential structure for antigen presentation in mouse CHS responses. An interesting remaining question is the role of iSALT in inflammatory skin dis- eases other than allergic contact dermatitis. In fact, in psoriasis, an iSALT-like structure has been reported in the skinlesions in humans74. Therefore, iSALT may serve as an important structure that supports the devel- opment of immune responses in other inflammatory skin diseases. iSALT and other tertiary lymphoid structures. Under some pathological circumstances such as infection, chronic inflammation and cancers, lymph node-like structures called tertiary lymphoid structures (TLSs) have been detected in various non-lymphoid tissues75. In the lung, for example, leukocyte clusters called induc- ible bronchial-associated lymphoid tissue (iBALT) are formed after pulmonary inflammation or infection76. iBALT can support the priming of naive B cells and T cells and also maintain memory B cell and T cell popu- lations. Thus, iBALTs are considered essential structures that support rapid and efficient immune responses in the lung against some pathogens. TLSs seem to play important roles in host protec- tion against external antigens and also against internal antigens in the development of pathogenic conditions such as autoimmune diseases and cancers75,77,78. The TLSs involved in skin immune responses have not yet been clearly defined, but in order to be considered bona fide TLSs, they should show several of the following characteristics: the existence of distinct T cell and B cell compartments, the presence of a follicular reticular cell (FRC) network, the expression of peripheral node addressin (PNAd)+, the presence of high endothelial venules (HEVs) and a lymphatic vasculature, and evi- dence of B cell class switching77. Most TLSs are not genetically programmed and do not develop postnatally. Instead, cytokines (lymphotoxin and TNF), lymphoid chemokines (CCL19, CCL21 and CXCL13) and recep- tor activator of nuclear factor-κB ligand (RANKL; also known as TNFSF11) play crucial roles in the develop- ment of TLSs. The characteristics of each leukocyte cluster are summarized in TABLe 2. On the basis of these structural criteria, iSALT may not be classified as a TLS because of the absence of B cells, naive T cells, HEVs and the FRC network and the involvement of lymphoid chemokines have not yet been described in iSALT. However, from a func- tional point of view, iSALT possesses the key feature of TLS in that it is an efficient site for effector T cell activation; some researchers thus use the term TLS for iSALT, focusing on this point78,79. Leukocyte clus- ters are also reported in the genital mucosa after HSV infection and have been named ‘memory lymphocyte clusters’80. They are mainly composed of CD11c+ MHC class II+ cells and CD4+ memory T cells and do not con- tain PNAd+ HEVs or lymphatic vessels. CD4+ memory T cells are recruited and maintained in the cluster through CCL5 produced by macrophages. In the mem- ory lymphocyte clusters, CD4+ T cells rarely recirculate into the blood and are critical for preventing the local reinfection of HSV. iSALT formation in human skin. It remains unclear whether iSALT is formed in human skin and supports T cell activation in an analogous manner to that seen in mice. Although iSALT-like structures (perivascular leukocyte infiltrations) are frequently observed by histo- logical examination in many inflammatory skin diseases, these may simply reflect the extravasation process of leu- kocytes, and it is not clear whether activation of effector T cells actually occurs in these structures. However, in some inflammatory skin diseases, this structure may serve as an iSALT by presenting antigens to effector T cells. In allergic contact dermatitis, DC–T cell clusters are found in the dermis and are accompanied by vesi- cle formation, a marker of inflammatory cytokine pro- duction in the epidermis above the cluster that suggest Table 1 | Putative APCs in the skin under different pathological conditions Disease model Putative antigen-presenting cells in the skin Contact hypersensitivity Dermal DCs20, mast cells56, blood endothelial cells61, radioresistant cells60 or LCs73,97 Graft versus host diseases LCs58 or keratinocytes63 Psoriasis Dermal DCs98 or LCs59 DC, dendritic cell; LC, Langerhans cell. Table 2 | Characteristics and comparison of leukocyte clusters Characteristic Type of leukocyte cluster iSALT Memory lymphocyte clusters iBALT Presence of HEVs, lymphatic vessels and FRC No No Yes Mediators involved in formation CXCL2, IL-1α and LTB4 CCL5 and CXCL9 Lymphotoxin, IL-17 , TNF, CCL19, CCL20 and CXCL13 Supports priming of naive T cells and B cells No No Yes Supports reactivation of effector T cells Yes Yes Yes Existence in human Unknown Unknown Yes CCL , CC-chemokine ligand; CXCL , CXC-chemokine ligand; FRC, follicular reticular network; HEV, high endothelial venule; iBALT, inducible bronchial-associated lymphoid tissue; iSALT, inducible skin-associated lymphoid tissue; LTB4, leukotriene B4; TNF, tumour necrosis factor. NATuRe RevIeWS | IMMuNoLogy R e v i e w s volume 19 | JANuARY 2019 | 27 that the activation of T cells occurs at the cluster20. In psoriasis, the existence of HEVs has been reported81, and clusters of DC-LAMP (also known as LAMP3)+ DCs and T cells have been detected in the dermis74 with abundant expression of CCL19 and CCL20 (ReFs82,83). This cluster disappears after treatment with TNF inhibitors74, sug- gesting the involvement of TNF in the maintenance of the cluster. iSALT-like structures with CXCL13+ cells have been reported in the skin lesions of patients with secondary syphilis infections69. In melanoma, clus- ters with TLS features, such as the existence of HEVs, T cells, B cells and mature DCs, have been detected in the extratumoural area and are associated with tumour regression or favourable overall survival in patients84,85. Lymphoid follicles are created in the skin lesions of cutaneous lupus erythematosus86 or Kimura disease87. Thus, although the functional importance of the leu- kocyte clusters or lymphoid follicles in the skin remains unclear, these structures may play important roles in the promotion or regulation of disease development (TABLe 3). Conclusions and perspectives The skin is a barrier organ that separates the body from the external environment. The skin consists of three main parts: the epidermis, dermis and subcuta- neous tissues. Each constituent has unique cells and structures, including immune cells and non-immune cells, which induce various immune responses to external stimuli in a coordinated manner. Substantial progress has been made in understanding the func- tions of the different cell types in the skin and the mechanisms that they use to communicate harmoni- ously. In particular, the understanding of the roles of non-immune cells, such as vascular cells and neurons, has shed light on the complexity of skin immunity and diseases. In addition, the identification of key loca- tions and/or structures, such as hair follicles, perivas- cular extravasation units and iSALT, has improved the understanding of cutaneous immune responses (Fig. 6). Moreover, it is now known that commensal and pathogenic bacteria directly regulate the func- tional properties of skin immune cells, which has pro- vided new paradigms that highlight the importance of host–microorganism interactions. Despite such progress, we still lack a comprehen- sive understanding of the skin in health and disease. Table 3 | Leukocyte clusters in human diseases in skin or genital mucosa Disease Characteristics of the clusters Refs Contact dermatitis Clusters of CD3+ cells and CD11c+ cells below epidermal vesicles 20 Psoriasis Existence of DC-L AMP+ DCs and T cells with CCL19 and CCL20 expression in the clusters 83 Secondary syphilis Existence of CXCL13+ fibroblast-like cells 69 HSV infection (skin) Variable numbers of CD30+ or CD56+ cells 99 HSV infection (genital mucosa) Existence of CCR5+CD4+ T cells contiguous to CD123 or DC-SIGN+ DCs 100 Chlamydia trachomatis infection (genital mucosa) Germinal centre formation 101 Kimura disease Germinalcentre formation 87 Cutaneous lupus erythematosus Existence of plasmacytoid DCs 86,102 CCL , CC-chemokine ligand; CCR5, CC-chemokine receptor 5; CXCL13, CXC-chemokine ligand 13; DC, dendritic cell; HSV, herpes simplex virus. Epidermis Dermis Hair follicles • Recruitment of immune cells (LCs and T cells) • Memory T cell responses (T RM cells) • Immune privilege (T reg cells) Postcapillary venules • Formation of iSALT (transient effector T cell activation) • Formation of perivascular extravasation units (neutrophil extravasation) Fig. 6 | Newly identified key locations and structures in skin immunity. Hair follicles serve as locations for immune cell recruitment and create structures for memory T cell response as well as immune privilege. Inducible skin-associated lymphoid tissue (iSALT) and perivascular extravasation units are formed around postcapillary venules, which induce the transient activation of effector T cells and neutrophil activation, respectively. LC, Langerhans cell; Treg cell, regulatory T cell; TRM cell, resident memory T cell. Kimura disease A chronic inflammatory disorder characterized by a painless lymphadenopathy or masses on head and neck regions. www.nature.com/nri R e v i e w s 28 | JANuARY 2019 | volume 19 We do not exactly understand which immune cells and/or non-immune cells interact with each other and at which immune response time points these interactions occur (for example, during the acute, chronic and resolution stages of inflammation); we also do not know the pre- cise anatomical locations of these interactions in the skin. Considering the fundamental differences between mouse and human skin is another important challenge. Evaluation of these points may lead to a breakthrough in the understanding of the immunological mechanisms of various cutaneous immune responses and healthy skin homeostasis. Published online 14 November 2018 1. Hoeffel, G. et al. Adult Langerhans cells derive predominantly from embryonic fetal liver monocytes with a minor contribution of yolk sac–derived macrophages. J. Exp. Med. 209, 1167–1181 (2012). 2. Ginhoux, F. et al. Langerhans cells arise from monocytes in vivo. Nat. Immunol. 7, 265–273 (2006). 3. Gebhardt, T. et al. Memory T cells in nonlymphoid tissue that provide enhanced local immunity during infection with herpes simplex virus. Nat. Immunol. 10, 524–530 (2009). 4. Masopust, D. et al. Dynamic T cell migration program provides resident memory within intestinal epithelium. J. Exp. Med. 207, 553–564 (2010). 5. Wakim, L. M., Woodward-Davis, A. & Bevan, M. J. Memory T cells persisting within the brain after local infection show functional adaptations to their tissue of residence. Proc. Natl Acad. Sci. USA 107, 17872–17879 (2010). 6. Mackay, L. K. 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 109, 7037–7042 (2012). 7. Wu, T. et al. Lung-resident memory CD8 T cells (TRM) are indispensable for optimal cross-protection against pulmonary virus infection. J. Leukoc. Biol. 95, 215–224 (2014). 8. Matsui, T. & Amagai, M. Dissecting the formation, structure and barrier function of the stratum corneum. Int. Immunol. 27, 269–280 (2015). 9. Egawa, G. & Kabashima, K. Multifactorial skin barrier deficiency and atopic dermatitis: essential topics to prevent the atopic march. J. Allergy Clin. Immunol. 138, 350–358.e351 (2016). 10. Belkaid, Y. & Tamoutounour, S. The influence of skin microorganisms on cutaneous immunity. Nat. Rev. Immunol. 16, 353–366 (2016). 11. Byrd, A. L., Belkaid, Y. & Segre, J. A. The human skin microbiome. Nat. Rev. Microbiol. 16, 143–155 (2018). 12. Tong, P. L. et al. The skin immune atlas: three- dimensional analysis of cutaneous leukocyte subsets by multiphoton microscopy. J. Invest. Dermatol. 135, 84–93 (2015). 13. Lämmermann, T. et al. Neutrophil swarms require LTB4 and integrins at sites of cell death in vivo. Nature 498, 371–375 (2013). 14. Jain, R., Tikoo, S., Egawa, G. & Weninger, W. in Encyclopedia of Immunobiology Vol. 3 (ed. Ratcliffe, M.) 493–504 (Elsevier, 2016). 15. Tamoutounour, S. et al. Origins and functional specialization of macrophages and of conventional and monocyte-derived dendritic cells in mouse skin. Immunity 39, 925–938 (2013). 16. Wolf, K., Müller, R., Borgmann, S., Bröcker, E.-B. & Friedl, P. Amoeboid shape change and contact guidance: T-lymphocyte crawling through fibrillar collagen is independent of matrix remodeling by MMPs and other proteases. Blood 102, 3262–3269 (2003). 17. Tomura, M. 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. 120, 883–893 (2010). 18. Kashem, S. W. et al. Nociceptive sensory fibers drive interleukin-23 production from CD301b+ dermal dendritic cells and drive protective cutaneous immunity. Immunity 43, 515–526 (2015). 19. Riol-Blanco, L. et al. Nociceptive sensory neurons drive interleukin-23-mediated psoriasiform skin inflammation. Nature 510, 157–161 (2014). 20. Natsuaki, Y. et al. Perivascular leukocyte clusters are essential for efficient activation of effector T cells in the skin. Nat. Immunol. 15, 1064–1069 (2014). This study shows that perivascular leukocyte clusters including macrophages and DCs are essential structures for effector T cell activation in the skin and proposes the concept of iSALT. 21. Carmi-Levy, I., Homey, B. & Soumelis, V. A modular view of cytokine networks in atopic dermatitis. Clin. Rev. Allergy Immunol. 41, 245–253 (2011). 22. Nestle, F. O., Di Meglio, P., Qin, J.-Z. & Nickoloff, B. J. Skin immune sentinels in health and disease. Nat. Rev. Immunol. 9, 679–691 (2009). 23. Nagao, K. et al. Stress-induced production of chemokines by hair follicles regulates the trafficking of dendritic cells in skin. Nat. Immunol. 13, 744–752 (2012). This is the first study to show the importance of hair follicles for immune cell trafficking from dermis to the epidermis. 24. Wollenberg, A., Kraft, S., Hanau, D. & Bieber, T. Immunomorphological and ultrastructural characterization of Langerhans cells and a novel, inflammatory dendritic epidermal cell (IDEC) population in lesional skin of atopic eczema. J. Invest. Dermatol. 106, 446–453 (1996). 25. Liu, Z. et al. Visualization of T cell-regulated monocyte clusters mediating keratinocyte death in acquired cutaneous immunity. J. Invest. Dermatol. 138, 1328–1337 (2018). This study demonstrates that monocytes cluster around hair follicles after hapten painting to the skin. 26. Paus, R. et al. The hair follicle and immune privilege. J. Investig. Dermatol. Symp. Proc. 8, 188–194 (2003). 27. Kang, H. et al. Hair follicles from alopecia areata patients exhibit alterations in immune privilege- associated gene expression in advance of hair loss. J. Invest. Dermatol. 130, 2677–2680 (2010). 28. Adachi, T. et al. Hair follicle–derived IL-7 and IL-15 mediate skin-resident memory T cell homeostasis and lymphoma. Nat. Med. 21, 1272–1279 (2015). 29. Collins, N. et al. Skin CD4+ memory T cells exhibit combined cluster-mediated retention and equilibration with the circulation. Nat. Commun. 7, 11514 (2016). This study shows that leukocyte clusters around hair follicles may serve as the structures for memory T cell activation after hapten application or HSV infection. 30. Ali, N. et al. Regulatory T cells in skin facilitate epithelial stem cell differentiation. Cell 169, 1119–1129.e1111 (2017). 31. Mattii, M. et al. Sebocytes contribute to skin inflammation by promoting the differentiation of T helper 17 cells. Br. J. Dermatol. 178, 722–730 (2018). 32. Nakahigashi, K. et al. PGD2 induces eotaxin-3 via PPARγ from sebocytes: a possible pathogenesis of eosinophilic pustular folliculitis. J. Allergy Clin. Immunol. 129, 536–543 (2012).33. Lai, Y. et al. Commensal bacteria regulate Toll-like receptor 3–dependent inflammation after skin injury. Nat. Med. 15, 1377–1382 (2009). 34. Linehan, J. L. et al. Non-classical immunity controls microbiota impact on skin immunity and tissue repair. Cell 172, 784–796 (2018). 35. Grice, E. A. et al. A diversity profile of the human skin microbiota. Genome Res. 18, 1043–1050 (2008). 36. Egawa, N., Egawa, K., Griffin, H. & Doorbar, J. Human papillomaviruses; epithelial tropisms, and the development of neoplasia. Viruses 7, 3863–3890 (2015). 37. Egawa, G. et al. Intravital analysis of vascular permeability in mice using two-photon microscopy. Sci. Rep. 3, 1932 (2013). 38. Abtin, A. et al. Perivascular macrophages mediate neutrophil recruitment during bacterial skin infection. Nat. Immunol. 15, 45–53 (2014). 39. Szallasi, A., Cortright, D. N., Blum, C. A. & Eid, S. R. The vanilloid receptor TRPV1: 10 years from channel cloning to antagonist proof-of-concept. Nat. Rev. Drug Discov. 6, 357–372 (2007). 40. van der Fits, L. et al. Imiquimod-induced psoriasis-like skin inflammation in mice is mediated via the IL-23/ IL-17 axis. J. Immunol. 182, 5836–5845 (2009). 41. Feng, J. et al. Sensory TRP channels contribute differentially to skin inflammation and persistent itch. Nat. Commun. 8, 980 (2017). 42. Nakamizo, S. & Egawa, G. in Immunology of the Skin (ed. Kabashima, K.) 227–238 (Springer, 2016). 43. Weisberg, S. P. et al. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Invest. 112, 1796–1808 (2003). 44. Zhang, L.-J. et al. Dermal adipocytes protect against invasive Staphylococcus aureus skin infection. Science 347, 67–71 (2015). 45. Egawa, G., Miyachi, Y. & Kabashima, K. Identification of perivascular adipose tissue in the mouse skin using two-photon microscopy. J. Dermatol. Sci. 70, 139–140 (2013). 46. Gao, Y. J., Lu, C., Su, L. Y., Sharma, A. & Lee, R. Modulation of vascular function by perivascular adipose tissue: the role of endothelium and hydrogen peroxide. Br. J. Pharmacol. 151, 323–331 (2007). 47. Rajsheker, S. et al. Crosstalk between perivascular adipose tissue and blood vessels. Curr. Opin. Pharmacol. 10, 191–196 (2010). 48. Kubo, A., Nagao, K., Yokouchi, M., Sasaki, H. & Amagai, M. External antigen uptake by Langerhans cells with reorganization of epidermal tight junction barriers. J. Exp. Med. 206, 2937–2946 (2009). 49. Dress, R. J., Wong, A. Y. & Ginhoux, F. Homeostatic control of dendritic cell numbers and differentiation. Immunol. Cell Biol. 96, 463–476 (2018). 50. Guilliams, M. et al. Unsupervised high-dimensional analysis aligns dendritic cells across tissues and species. Immunity 45, 669–684 (2016). 51. Schlitzer, A., McGovern, N. & Ginhoux, F. Dendritic cells and monocyte derived cells: two complementary and integrated functional systems. Semin. Cell Dev. Biol. 41, 9–22 (2017). 52. Honda, T. et al. Tuning of antigen sensitivity by T cell receptor-dependent negative feedback controls T cell effector function in inflamed tissues. Immunity 40, 235–247 (2014). This study shows a regulatory mechanism of effector T cell motility and its activation status in the skin. 53. Krummel, M. F., Bartumeus, F. & Gerard, A. T cell migration, search strategies and mechanisms. Nat. Rev. Immunol. 16, 193–201 (2016). 54. Egawa, G. et al. In vivo imaging of T cell motility in the elicitation phase of contact hypersensitivity using two-photon microscopy. J. Invest. Dermatol. 131, 977–979 (2011). 55. Matheu, M. P. et al. Imaging of effector memory T cells during a delayed-type hypersensitivity reaction and suppression by Kv1.3 channel block. Immunity 29, 602–614 (2008). 56. Dudeck, J. et al. Mast cells acquire MHCII from dendritic cells during skin inflammation. J. Exp. Med. 214, 3791–3811 (2017). 57. Miyake, K. et al. Trogocytosis of peptide-MHC class II complexes from dendritic cells confers antigen- presenting ability on basophils. Proc. Natl Acad. Sci. USA 114, 1111–1116 (2017). 58. Bennett, C. L. et al. Langerhans cells regulate cutaneous injury by licensing CD8 effector cells recruited to the skin. Blood 117, 7063–7069 (2011). 59. Kim, J. H. et al. CD1a on Langerhans cells controls inflammatory skin disease. Nat. Immunol. 17, 1159–1166 (2016). This study shows that LCs play crucial roles in the induction of allergic reactions to urushiol as well as in psoriasis via their expression of CD1a. 60. Ono, S., Honda, T. & Kabashima, K. Requirement of MHC class I on radioresistant cells for granzyme B expression from CD8+ T cells in murine contact hypersensitivity. J. Dermatol. Sci. 90, 98–101 (2018). 61. Kish, D. D., Volokh, N., Baldwin, W. M. 3rd & Fairchild, R. L. Hapten application to the skin induces an inflammatory program directing hapten-primed effector CD8 T cell interaction with hapten-presenting endothelial cells. J. Immunol. 186, 2117–2126 (2011). 62. Gaspari, A. A. & Katz, S. I. Induction and functional characterization of class II MHC (Ia) antigens on NATuRe RevIeWS | IMMuNoLogy R e v i e w s volume 19 | JANuARY 2019 | 29 murine keratinocytes. J. Immunol. 140, 2956–2963 (1988). 63. Kim, B. S. et al. Keratinocytes function as accessory cells for presentation of endogenous antigen expressed in the epidermis. J. Invest. Dermatol. 129, 2805–2817 (2009). 64. Krummel, M. F., Heath, W. R. & Allison, J. Differential coupling of second signals for cytotoxicity and proliferation in CD8+ T cell effectors: amplification of the lytic potential by B7. J. Immunol. 163, 2999–3006 (1999). 65. Streilein, J. W. Skin-associated lymphoid tissues (SALT): origins and functions. J. Invest. Dermatol. 80 (Suppl.), 12–16 (1983). 66. Streilein, J. W. Circuits and signals of the skin- associated lymphoid tissues (SALT). J. Invest. Dermatol. 85, S10–S13 (1985). 67. Sontheimer, R. Perivascular dendritic macrophages as immunobiological constituents of the human dermal microvascular unit. J. Invest. Dermatol. 93, S96–S101 (1989). 68. Honda, T. & Kabashima, K. Novel concept of iSALT (inducible skin-associated lymphoid tissue) in the elicitation of allergic contact dermatitis. Proc. Jpn. Acad. 92, 20–28 (2016). 69. Kogame, T. et al. Possible inducible skin-associated lymphoid tissue (iSALT)-like structures with CXCL13+ fibroblast-like cells in secondary syphilis. Br. J. Dermatol. 177, 1737–1739 (2017). 70. Sawada, Y. et al. Resolvin E1 inhibits dendritic cell migration in the skin and attenuates contact hypersensitivity responses. J. Exp. Med. 212, 1921–1930 (2015). 71. Kashem, S. W., Haniffa, M. & Kaplan, D. H. Antigen- presenting cells in the skin. Annu. Rev. Immunol. 35, 469–499 (2017). 72. Okada, T., Takahashi, S., Ishida, A. & Ishigame, H. In vivo multiphoton imaging of immune cell dynamics. Pflugers Arch. 468, 1793–1801 (2016). This study shows the in vivo dynamics of skin DC subsets in iSALT by multiphoton microscopy. 73. 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. 125, 1154–1156.e1152 (2010). 74. Zaba, L. C. et al. Amelioration of epidermal hyperplasia by TNF inhibition is associated with reduced Th17 responses. J. Exp. Med. 204, 3183–3194 (2007). 75. Pitzalis, C., Jones, G. W., Bombardieri, M. & Jones, S. A. Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat. Rev. Immunol. 14, 447–462 (2014). 76. Randall, T. D. Bronchus-associated lymphoid tissue (BALT) structure and function. Adv. Immunol. 107, 187–241 (2010). 77. Dieu-Nosjean, M. C., Goc, J., Giraldo, N. A., Sautes-Fridman, C. & Fridman, W. H. Tertiary lymphoid structures in cancer and beyond. Trends Immunol. 35, 571–580 (2014). 78. Colbeck, E. J., Ager, A., Gallimore, A. & Jones, G. W. Tertiary lymphoid structures in cancer: drivers of antitumor immunity, immunosuppression, orbystander sentinels in disease? Front. Immunol. 8, 1830 (2017). 79. Neyt, K., Perros, F., GeurtsvanKessel, C. H., Hammad, H. & Lambrecht, B. N. Tertiary lymphoid organs in infection and autoimmunity. Trends Immunol. 33, 297–305 (2012). 80. Iijima, N. & Iwasaki, A. A local macrophage chemokine network sustains protective tissue-resident memory CD4 T cells. Science 346, 93–98 (2014). This study shows that leukocyte clusters in genital mucosa after HSV infection are essential structures where activation of memory T cells is induced for the elimination of HSV. 81. Lowe, P. M. et al. The endothelium in psoriasis. Br. J. Dermatol. 132, 497–505 (1995). 82. Mitsui, H. et al. Combined use of laser capture microdissection and cDNA microarray analysis identifies locally expressed disease-related genes in focal regions of psoriasis vulgaris skin lesions. J. Invest. Dermatol. 132, 1615–1626 (2012). 83. Kim, T. G. et al. Dermal clusters of mature dendritic cells and T cells are associated with the CCL20/CCR6 chemokine system in chronic psoriasis. J. Invest. Dermatol. 134, 1462–1465 (2014). 84. Martinet, L. et al. High endothelial venules (HEVs) in human melanoma lesions: major gateways for tumor-infiltrating lymphocytes. Oncoimmunology 1, 829–839 (2012). 85. Ladanyi, A. et al. Density of DC-LAMP+ mature dendritic cells in combination with activated T lymphocytes infiltrating primary cutaneous melanoma is a strong independent prognostic factor. Cancer Immunol. Immunother. 56, 1459–1469 (2007). 86. Arps, D. P. & Patel, R. M. Lupus profundus (panniculitis): a potential mimic of subcutaneous panniculitis-like T cell lymphoma. Arch. Pathol. Lab. Med. 137, 1211–1215 (2013). 87. Kung, I. T., Gibson, J. B. & Bannatyne, P. M. Kimura’s disease: a clinico-pathological study of 21 cases and its distinction from angiolymphoid hyperplasia with eosinophilia. Pathology 16, 39–44 (1984). 88. Murata, T. et al. Transient elevation of cytoplasmic calcium ion concentration at a single cell level precedes morphological changes of epidermal keratinocytes during cornification. Sci. Rep. 8, 6610 (2018). 89. Honda, T., Egawa, G., Grabbe, S. & Kabashima, K. Update of immune events in the murine contact hypersensitivity model: toward the understanding of allergic contact dermatitis. J. Invest. Dermatol. 133, 303–315 (2013). 90. Kaplan, D. H., Igyártó, B. Z. & Gaspari, A. A. Early immune events in the induction of allergic contact dermatitis. Nat. Rev. Immunol. 12, 114–124 (2012). 91. Topalian, S. L. et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366, 2443–2454 (2012). 92. Boissonnas, A. et al. CD8+ tumor-infiltrating T cells are trapped in the tumor-dendritic cell network. Neoplasia 15, 85–94 (2013). 93. Engelhardt, J. J. et al. Marginating dendritic cells of the tumor microenvironment cross-present tumor antigens and stably engage tumor-specific T cells. Cancer Cell 21, 402–417 (2012). 94. Broz, M. L. et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell 26, 638–652 (2014). This study identifies the cutaneous DC subset responsible for antigen presentation in the skin in melanoma. 95. Gardner, A. & Ruffell, B. Dendritic cells and cancer immunity. Trends Immunol. 37, 855–865 (2016). 96. Gordon, S. R. et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature 545, 495–499 (2017). 97. Noordegraaf, M., Flacher, V., Stoitzner, P. & Clausen, B. E. Functional redundancy of Langerhans cells and Langerin+ dermal dendritic cells in contact hypersensitivity. J. Invest. Dermatol. 130, 2752–2759 (2010). 98. Wohn, C. et al. Langerinneg conventional dendritic cells produce IL-23 to drive psoriatic plaque formation in mice. Proc. Natl Acad. Sci. USA 110, 10723–10728 (2013). 99. Leinweber, B., Kerl, H. & Cerroni, L. Histopathologic features of cutaneous herpes virus infections (herpes simplex, herpes varicella/zoster): a broad spectrum of presentations with common pseudolymphomatous aspects. Am. J. Surg. Pathol. 30, 50–58 (2006). 100. Zhu, J. et al. Persistence of HIV-1 receptor-positive cells after HSV-2 reactivation is a potential mechanism for increased HIV-1 acquisition. Nat. Med. 15, 886–892 (2009). 101. Kiviat, N. B. et al. Endometrial histopathology in patients with culture-proved upper genital tract infection and laparoscopically diagnosed acute salpingitis. Am. J. Surg. Pathol. 14, 167–175 (1990). 102. Farkas, L., Beiske, K., Lund-Johansen, F., Brandtzaeg, P. & Jahnsen, F. L. Plasmacytoid dendritic cells (natural interferon- α/β-producing cells) accumulate in cutaneous lupus erythematosus lesions. Am. J. Pathol. 159, 237–243 (2001). Acknowledgements This work was supported by grants from the Japan Society for the Promotion of Science KAKENHI (JP15K09766, JP15H05096 (to T.H.) and 263395 (to K.K)), Grants-in-Aid for Scientific Research (15H05790, 15H1155 and 15K15417 to K.K.) and the Japan Agency for Medical Research and Development (AMED) (16ek0410011h0003 and 16he0902003h0002 to K.K.). The authors thank A. Hayday of the King’s College London School of Medicine, London, UK, and E. Epstein Jr of PellePharm for the critical reading of the manuscript. Author contributions All authors contributed to the discussion of the content of the article. G.E. and T.H. also contributed to researching data and the writing of the article. K.K. and F.G. also contributed to the review and editing of the manuscript. Competing interests The authors declare no competing interests. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Reviewer information Nature Reviews Immunology thanks B. Malissen and the other anonymous reviewer(s) for their contribution to the peer review of this work. www.nature.com/nri R e v i e w s 30 | JANuARY 2019 | volume 19 The immunological anatomy of the skin Stratum corneum Immunological anatomy of the epidermis Monocyte interaction with hair follicles. T cell interaction with hair follicles. Immune modulation by sebaceous glands. Influence of the skin microbiota. Immune responses in the dermis Immune modulation by dermal blood vessels. Neuro-immune interactions in the skin. Contact hypersensitivity responses Immunity in subcutaneous adipose tissue Skin anatomy and induction of adaptive immunity Antigen presentation by cutaneous APCs. Antigen presentation in skin neoplasms iSALT formation around postcapillary venules. iSALT and other tertiary lymphoid structures. iSALT formation in human skin. Conclusions and perspectives Acknowledgements Fig. 1 Physical and immunological barrier of the skin. Fig. 2 A specific contribution of postcapillary venules in cutaneous immunity. Fig. 3 Neuro-immune interactions and perivascular adipose tissue in the skin. Fig. 4 Penetration of hapten and proteins into the skin. Fig. 5 A schematic view of the sensitization and elicitation phases of contact hypersensitivity. Fig. 6 Newly identified key locations and structures in skin immunity. Table 1 Putative APCs in the skin under different pathological conditions. Table 2 Characteristics and comparison of leukocyte clusters. Table 3 Leukocyte clusters in human diseases in skin or genital mucosa.