Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

REviEwS

T cells and the skin: from protective


immunity to inflammatory skin
disorders
Allen W. Ho and Thomas S. Kupper*
Abstract | Skin is our primary interface with the environment, and T cells are crucial for
orchestrating host immune responses against pathogenic microorganisms at this site.
Effective skin immune responses require the generation of antigen-specific effector T cells,
which home to cutaneous sites of injury or infection. Long-lasting immunity against future immune
challenges is mediated by memory T cells. Among the memory T cells found in skin are both
recirculating cells that transit between skin and blood and tissue-resident memory T (TRM) cells,
which remain in skin for long periods of time and mediate durable protective immunity. These
TRM cells also appear to drive many inflammatory diseases of skin. Here, we consider how a better
understanding of cutaneous T cell responses can aid in the development of effective new
therapies for immune-mediated cutaneous diseases.

Barrier tissues like skin are under constant siege Skin T cell responses
from an environment in which physical, chemical Generation of effector T cell responses in skin. Skin is
and biological threats are abundant and ubiquitous. a formidable physical barrier, and many of its intrinsic
Experimental studies of immune cells isolated from cells contribute to this property — keratinocytes differ-
human blood or from secondary lymphoid tissues in entiate and form the impermeable stratum corneum6,
mice were once thought to be sufficient to provide a while fibroblasts produce collagens, elastin and other
comprehensive understanding of adaptive immune glycoproteins that give the dermis its toughness and
memory. However, recent findings have challenged resilience7. Additionally, nearly every type of skin cell
this paradigm. It has been estimated that at least as can, upon stimulation, produce chemokines, cytokines
many memory T cells exist in peripheral non-lymphoid and other inflammatory mediators that activate cells of
tissues as in secondary lymphoid tissues at any given the innate and adaptive immune systems and promote
time, either as transient or more permanent residents1. leukocyte recruitment from blood4,5,8. Trauma to the epi-
Furthermore, these tissue-dwelling T cells have very dermis, infection or exposure to ultraviolet radiation can
different properties from the T cells found in blood activate skin cells, typically via the NF-κB-dependent
and lymph nodes. In this Review, we focus on the and MAP kinase-dependent pathways4,5,8–10. Innate
T cell responses that develop in response to infections immune activation of dermal DCs leads to their matu­
or following antigen encounter in the skin2–4. Adult ration into powerful antigen-presenting cells. Mature
human skin is home to abundant populations of mem- DCs migrate through afferent lymphatics to the draining
ory αβ T cells, dendritic cells (DCs) and macrophages, lymph nodes, where they can activate naive T cells or
as well as lesser numbers of natural killer (NK) cells, recirculating central memory T cells. Activated DCs can
γδ T cells and innate lymphoid cells (ILCs)4,5 (Fig. 1). also activate recruited or tissue-resident T cells in the
Although effector and memory T cells coordinate skin11. Episodic recruitment of T cells and other leuko-
cutaneous immune responses against microorganisms cytes into inflamed or injured skin occurs many times
and cancer, aberrant or inappropriate T cell activation each day5,12 (Fig. 1).
Department of Dermatology, against innocuous or auto-antigens can lead to chronic Adult skin contains large populations of memory
Brigham and Women’s inflammatory disorders of skin. Here, we discuss adap- T cells that are phenotypically, functionally and clonally
Hospital, Harvard Medical
School, Boston, MA, USA.
tive immune responses in the skin, particularly those diverse13–15. Memory T cells are, by definition, derived
mediated by T cells that dwell in skin for varying from naive T cells that have undergone multiple rounds
*e-mail: tkupper@
bwh.harvard.edu periods of time, and discuss the role of these cells in of clonal expansion16. Skin-tropic effector T cell clones
https://doi.org/10.1038/ several immune-mediated dermatological diseases are generated from naive T cell precursors that have
s41577-019-0162-3 afflicting humans. been activated by antigen-presenting DCs in lymph

Nature Reviews | Immunology


Reviews

CD8+ DETC
LC TRM cell (mouse)
Stratum
corneum
Stratum
granulosum

Epidermis
Epidermis
Stratum
spinosum

Stratum Melanocyte
basale
CD4+
γδ T cell TRM cell CD8+
TRM cell
Papillary ILC
dermis DC

NK cell
Dermis
Macrophage

Fibroblast
Reticular
dermis
Recirculating
memory CD4+
T cell
Recirculating Afferent
Blood memory CD8+
vessel T cell lymphatic
vessel

Fig. 1 | The structure of the skin contributes to its immune functions. The epidermis is composed of layers of
increasingly well-differentiated keratinocytes, while the dermis includes superficial (papillary) and deep (reticular) layers.
Cutaneous immunology has benefited heavily from the study of murine skin as a model organism; however, key differences
between human and mouse skin exist. The most prevalent immune cell types that populate human epidermis are
Langerhans cells (LCs) and CD8+ tissue-resident memory T (TRM) cells. The murine epidermis contains dendritic epidermal
T cells (DETCs), which are absent in human epidermis. Both the mouse and human dermis are populated by dendritic cells
(DCs), macrophages, innate lymphoid cells (ILCs), natural killer (NK) cells and CD8+ TRM cells. The dermis also provides
structural support for the skin and, as such, consists of fibroblasts and connective tissue. In murine dermis, γδ T cells play a
prominent role in the production of IL-17 , while in human skin αβ T cells are responsible. The functional coordination of
these cellular mediators results in effective immune responses and a subsequent return to immune homeostasis.

nodes that drain the skin11,14. Clonally expanded effec- Generation of skin-homing T cells. It has been more
tor T cells generated under these conditions traffic to than 20 years since the initial observation that anti-
skin, accumulating in highest abundance at the site of gen encountered through skin leads to the generation
initial antigen encounter (Fig. 2). However, these cells of skin-homing T cells, whereas antigen encountered
also accumulate in non-inflamed skin, albeit in lower through the gastrointestinal tract generates gut-homing
numbers17. The effector T cell populations that are gen- T cells23–25. Given that the scope of this Review is lim-
erated during an acute immune response are far more ited to cutaneous T cell immunity, we focus on the
numerous than the memory T cell populations that generation of skin-homing T cells. While the features
survive long term in the skin, but the precise relation- in the tissue-draining lymph node microenvironment
ship between these populations is unclear18. One model that imprint T cells with tissue-homing properties are
suggests that memory T cells are a distinct lineage that not completely understood, many key changes appear
derive from an early activated T cell subpopulation within the first few T cell divisions26. In skin-draining
without going through effector T cell expansion18,19. By lymph nodes, uniquely glycosylated ligands for
contrast, another model suggests that memory T cells E-selectin and P-selectin (including cutaneous lympho­
have gone through considerable clonal expansion and cyte antigen (CLA)) are upregulated by activated
derive stochastically from a subset of surviving effector T cells in association with α(1,3) fucosyltransferases5,26.
T cells3,20,21. The heterogeneity of memory T cells bears In addition, this same population of activated T cells
emphasizing, not just with regards to their effector func- acquires expression of a set of chemokine receptors,
tions and cytokine profiles but also with regards to their including CC-chemokine receptor 4 (CCR4), CCR8
trafficking properties22. and CCR10 (ref.5). These E-selectin ligands in concert

www.nature.com/nri
Reviews

Priming phase Effector phase Memory phase


Lymphatic (3–10 days) (>25 days)

↓ T-bet
S1PR1, ↓ EOMES
CCR7 CD8+
Memory ↓ S1PR1
Circulating TRM cell
precursor ↓ SELL
TM cell Teff cell Teff cell TGFβ,
IL-15 ↑ RUNX3
↑ FAB4/FAB5
CLA ↑ CD69
↑ CD103
APC ↑ BCL-2
CD4+
↑ HOBIT
ESL, TRM cell
PSL, APC (mouse)
Activated T cell ↑ BLIMP1
CCR4,
Secondary CCR8, Pathogens (mouse)
lymphoid tissue CCR10

Skin

Blood

Fig. 2 | The generation and maintenance of cutaneous tissue-resident memory T cells. Following microbial invasion,
antigen-presenting cells (APCs) migrate from the site of insult to the secondary lymphoid tissues via afferent lymphatic
vessels. T cell priming occurs within the secondary lymphoid tissue, resulting in the generation of effector T cells (Teff cells,
which include memory T (TM) cell precursors) that travel to the site of inflammation. This pool of Teff cells eradicates the
infection, concluding the effector phase of an immune response, which typically lasts 3–10 days. Following microbial
clearance, the majority of Teff cells die, while subsets of T cells become recirculating memory cells or are maintained in the
tissue as tissue-resident memory T (TRM) cells. Recirculating memory T cells leave skin in response to gradients of
sphingosine-1-phosphate and CC-chemokine ligand 21 (CCL21), which bind to sphingosine-1-phosphate receptor 1 (S1PR1)
and CC-chemokine receptor 7 (CCR7), respectively. The generation of TRM cells results from a specific transcriptional
signature that has been characterized. TM cells are defined as cells that persist in the absence of inflammation or infection,
usually >25 days after the original exposure. BLIMP1, B lymphocyte-induced maturation protein 1; CL A , cutaneous
lymphocyte antigen; ESL , E-selectin; HOBIT, homologue of BLIMP1 in T cells; PSL , P-selectin; RUNX3, runt-related
transcription factor 3.

with skin-specific chemokine receptors and leukocyte CXC-chemokine receptor 3 (CXCR3), play an inte-
integrins allow for the tethering and arrest of these gral role in cutaneous T cell infiltration in this setting.
T cells in dermal post-capillary venules, and this results Inflamed skin expresses high levels of CXC-chemokine
in their extravasation into tissue5 (Fig. 2). Once on the ligand 9 (CXCL9) and CXCL10, which leads to the
abluminal side of the vessel, T cells respond to chemo- recruitment of CXCR3+ T cells to the sites of cutaneous
tactic gradients within the skin and migrate to the site inflammation32–36. However, CXCR3 is not absolutely
of injury or infection5. Vitamins A and D have been required for this process, as CXCR3-deficient CD8+
shown to regulate T cell homing to gut and cutane- T cells can be found in the skin during cutaneous vac-
ous tissues, respectively, via modulation of chemokine cinia virus (VACV) infection36, suggesting that CXCR3-
receptors and selectins or vascular adhesion molecule independent recruitment mechanisms also exist. In
ligands27,28. However, while the role of vitamin A in addition, although the recruitment of effector CD8+
imprinting gut T cell homing by inducing β7 integrin T cells to the lung and vagina requires IFNγ-producing
and CCR9 is broadly accepted, the role of vitamin D CD4+ T cells35,37, virus-specific tissue-resident memory T
in imprinting T cells with a skin-homing programme (TRM) cells develop and persist in the skin in the absence
is more controversial. The active metabolite of vitamin of CD4+ T cells17.
D3, 1α,25-dihydroxyvitamin D3, was shown to induce Skin infection preferentially generates skin-homing
CCR10 expression in human T cells and to a much lower effector T cells in draining lymph nodes, but there is evi-
degree in mouse T cells29. However, only a minority of dence for leakiness in the process. After VACV infec-
skin-homing T cells bear CCR10, and T cell expression tion of skin, CD8+ T cells upregulate E-selectin ligand
of the skin-homing chemokine CCR8 is independent of within five cell divisions, but a subset of proliferating
vitamin D; rather, keratinocyte-derived soluble factors CD8+ cells exit the lymph node after three cell divisions
lead to T cell expression of CCR8 (ref.30). Furthermore, and enter the mesenteric lymph nodes, where they
1α,25-dihydroxyvitamin D3 inhibits expression of CLA upregulate gut-homing molecules and are later detected
and, in parallel, functional E-selectin ligand, resulting in as α4β7 integrin-positive memory T cells in the gut26.
less skin infiltration of effector CD4+ T cells31. Although Blocking this early exit of proliferating cells from lymph
skin-homing molecules on memory T cells are essen- nodes using FTY720 (fingolimod) (an antagonist of
tial for entry into non-inflamed skin, active inflam- sphingosine-1-phosphate receptor 1 (S1PR1)) abrogates
mation makes T cell entry into skin more permissive. this process entirely26,38. Thus, the adaptive immune sys-
Signalling via G protein-coupled receptors, including tem hedges its bets by generating populations of effector

Nature Reviews | Immunology


Reviews

T cells with more than one tissue tropism, allowing for Non-recirculating T RM  cells in human skin were
the possibility that the pathogen will be encountered mostly CD4 + and CD69 + (ref. 15) . CD8 + T RM  cells
at a different environmental interface in the future. were mainly epidermal and expressed CD103, a ligand
Additional work done in a VACV skin infection model for E-cadherin. A more rapidly recirculating popula-
showed that, while most CD8+ effector T  cells are tion of T cells that expressed CLA, CCR4, L-selectin
recruited to inflamed skin, other CD8+ effector T cells and CCR7 were identified as skin TCM cells, whereas a
enter uninflamed skin throughout the body with similar more slowly recirculating population of skin T cells that
kinetics17. The size of the effector CD8+ T cell population expressed CCR7 (but not L-selectin) were termed migra-
in the skin peaks at 10 days after infection and declines tory memory T (TMM) cells, and it was proposed that
sharply thereafter17,39. Transcriptional profiling suggests these cells may be the direct precursors of TRM cells. As
that the effector T cell gene expression profile changes to TMM cells clearly recirculate, this may explain why multi-
a TRM cell profile at approximately day 25 post-challenge, ple discrete (that is, TRM cell-containing) skin lesions can
which then appears to persist indefinitely39,40 (Fig. 2). develop in both benign and malignant T cell-mediated
Memory T cells provide immunosurveillance and skin disorders15. It should be noted that TMM cells were
mediate a rapid response to future attacks from known described previously by other groups as recirculat-
pathogens. Nearly two decades ago, it was proposed that ing T cells that migrate from skin to lymph nodes via
two populations of circulating human memory T cells afferent lymphatics23,48.
exist, each with different trafficking properties41,42.
Central memory T (TCM) cells, like naive T cells, express Resident memory T cells. T RM  cells are defined as
CCR7 and CD62L and circulate between lymph nodes non-recirculating T cells that reside indefinitely in
and blood, while effector memory T (TEM) cells lack peripheral tissues; they have been identified in nearly
CCR7 but express tissue-homing addressins, allowing every normal and diseased tissue of the body, including
them to gain access to non-lymphoid peripheral tis- in malignant tumours2,3,22,55–59. Although the transcrip-
sues43. This paradigm, though still useful, continues to tional profiles of TRM cells isolated from brain, lung,
be modified substantially, as populations of T cells that skin and intestine differ slightly, there is a core gene
express either CCR7 or CD62L (but not both) have been expression signature that seems to be retained39,40,60–62.
demonstrated15,42. More recently, cells with CCR7 and Common features are the downregulation of genes
CD62L that also express the skin-homing molecules encoding S1PR1, EOMES and CD62L and the upreg-
CLA and CCR4 have been identified42,44, indicating that ulation of CD103 and various genes involved in adap-
cells with TCM cell markers can enter peripheral tissues. tive immune responses and lipid metabolism39,40 (Fig. 2).
These skin-homing TCM cells can give rise to TRM cells in CD69 appears to be nearly universally expressed on
skin16,44–46, and there is a subtype of cutaneous T cell lym- skin TRM cells, and it serves to interfere with the func-
phoma in which the malignant cells bear a skin-homing tion of S1PR1, thereby making TRM cells unresponsive
TCM cell phenotype42,47 and recirculate between skin and to the sphingosine-1-phosphate gradients that normally
blood. In addition, skin-homing TEM cells that express guide T cells to lymph nodes63. However, TRM cells in
CCR7 have been shown to enter skin and then exit skin other tissues do not express CD69 as faithfully as skin
via afferent lymphatics in response to CC-chemokine TRM cells64,65, suggesting that caution should be used
ligand 21 (CCL21) gradients15,48 (Fig. 2). in relying on this marker to define TRM cells. The tran-
scription factors that characterize TRM cells are also the
Skin-resident T cells. In 2001, it was demonstrated subject of some controversy — B lymphocyte-induced
that, after the resolution of systemic viral infection, maturation protein 1 (BLIMP; also known as PRDM1)
T cells are detected and persist for long periods of time and the homologue of BLIMP1 in T cells (HOBIT; also
in peripheral non-lymphoid tissues25,49–51. The term known as ZNF63) appear to be important for murine
skin-resident T cell first appeared in the early 2000s in TRM cells but not for human TRM cells. More recently,
reference to memory T cells found in lesions of fixed runt-related transcription factor 3 (RUNX3) was identi-
drug eruptions, which appear at the same location after fied as a necessary factor for tissue residence40,61,66 (Fig. 2).
repeated exposure to the same drug52. In 2004, Boyman It is unlikely that any single factor is sufficient to confer
et al. reported that non-lesional skin from patients tissue residency, and it is more likely that some redun-
with psoriasis (but not normal control skin) developed dancy of factors ensures this property; these include the
a psoriatic phenotype when grafted onto immuno­ above variables as well as dependence on lipid uptake
compromised mice53, suggesting that pathogenic T cells and metabolism, IL-15 and transforming growth
reside in normal-appearing adult skin. In 2006, a sem- factor-β (TGFβ)39,62,67–69.
inal report by Clark et al. demonstrated that there were TRM cells appear to survive, in part, by proliferating
roughly four times as many memory T cells in adult in situ at a low level45,46. A current paradigm holds that
human skin as there were in peripheral blood13. These TRM cells cannot leave tissue and recirculate and can be
skin T cells were heterogeneous and retained potent replaced by circulating T cells, with both circulating
effector functions, with different subsets producing TCM cells and TEM cells contributing16,44–46,70. Although
IFNγ, IL-17 or IL-13 overexpressing a regulatory T both TCM cells and TEM cells can become TRM cells16,44,70,
(Treg) cell phenotype13,54. More recently, the same group it is not known whether the TRM cell state represents ter-
described four distinct populations of memory T cells in minal differentiation. It was recently suggested that the
skin — a population of TCM cells, a population of migra- most proximal precursor to the TRM cells is the TMM cell15.
tory memory T cells and two populations of TRM cells15. Whether this differentiation is unidirectional or whether

www.nature.com/nri
Reviews

TRM cells can occasionally revert back to TMM cells and findings have also been extended to human skin. Even
exit the skin is currently unknown15,16. Although a single months after HSV-2 infection of human genital skin,
study demonstrated that the tissue microenvironment HSV-2-specific CD8+ T cells are present at the dermal–
could lead to the development of TCM cells or TEM cells epidermal junction and are in contact with keratino­
from gut-resident T cells upon reactivation71, the current cytes 83. Following cutaneous HSV-1 reactivation,
dogma based on more recent data is that TRM cells do not CD8+ T cells at the site of infection express the CD8αα
de-differentiate22,45,46,70. For example, cutaneous TRM cells homodimer rather than the CD8αβ heterodimer84. HSV-
appear to proliferate in situ but do not differentiate into 2-specific CD4+ T cells are retained at sites of HSV-2
other memory T cell subsets nor leave the skin to recir- reactivation for months after healing within the dermis85.
culate45. TRM cells from other epithelial tissues such as
the uterine mucosa and lung behave similarly46,70. It is T cells in skin: of mice and men. Most work on the
not yet known whether epigenetic changes occur in real-time generation of TRM cells in skin after microbial
TRM cells that limit their developmental plasticity, and or viral infection has been performed in mice86. By the
this p
­ ossibility is the subject of much current interest. time they reach adulthood, humans have lived for many
Antiviral CD8+ TRM cells can protect the host by years in a pathogen-rich world and have accumulated
directly killing virally infected cells72. In addition, memory T cell clones that inhabit both lymphoid tissue
TRM cell activation can induce a generalized antiviral and peripheral barrier tissues87,88. By contrast, labora-
state in skin, promoting the release of type I interferons tory mice are kept under carefully controlled specific
by keratinocytes and other skin cells56,73,74. As already pathogen-free (SPF) conditions and are typically exam-
mentioned, skin CD8+ TRM cells typically express CD103 ined at an early age — as such, they possess relatively few
and are localized to the epidermis, but CD103–CD8+ memory T cells and far more naive T cells86. In skin, mice
TRM cells have been reported32,33,75. By contrast, skin are born with two unique populations of γδ T cells in the
CD4+ TRM cells are less well characterized and appear to dermis and epidermis89. Epidermal γδ T cells, also known
localize in the papillary dermis13,15. CD4+ TRM cells spe- as dendritic epidermal T cells (DETCs), have a T cell
cific for herpes simplex virus (HSV) (T helper 1 (TH1) receptor (TCR) with a single antigenic specificity90 (Fig. 1).
cell-like), Leishmania spp. (TH1 cell-like) and Candida They are generated in the thymus during embryonic
albicans (TH17 cell-like) have all been described, and development and home to skin in utero91. Functionally,
their expression of CD69 is high, while their expression they are thought to play a role in wound healing and
of CD103 is variable76–78. They have been shown to either epidermal repair and may have an innate immune sig-
cluster near hair follicles or to be more evenly distrib- nalling function89,92–94. DETCs are sessile and do not
uted; these differences may be dependent on the model circulate out of the skin. Dermal γδ T cells are more
or pathogen15,76,79,80. Like CD8+ TRM cells, CD4+ TRM cells diverse with regard to antigen recognition and are
can be rapidly activated by pathogen re-challenge and important as a first line of defence for a subset of patho-
promote more rapid clearance of the pathogen from gens95. Parabiosis experiments suggest that migration of
skin76 (Fig. 2). There is also evidence that, in addition to dermal γδ cells resembles that of human skin TMM cells,
CD8+ TRM cells, local infection generates high levels of with slow kinetics of recirculation out of skin96. Before
CD4+ TRM cells at the site of infection and lower levels explicit or experimental antigenic challenge or infection,
throughout the entire skin, thus providing global protec- comparatively few αβ T cells exist in mouse skin, and
tion76. The question of whether TRM cells alone are suffi- many of these are recirculating Treg cells97. Unlike αβ
cient to protect against infection is also model-specific T cells, γδ T cells do not recognize peptides bound to
and pathogen-specific. For the VACV system, CD8+ MHC molecules, and different subsets appear to have
T cells alone without antibody or circulating T cells are different antigenic specificities94. In general, activating
sufficient for full protection17, whereas for Leishmania ligands for γδ T cells are incompletely characterized but
spp. infection, circulating T  cells are required to include host stress proteins like MICA and NKG2B98.
augment T RM  cell-mediated protective responses 77. In mouse skin, DETCs appear to respond to epidermal
Regardless of the specific infection, the posting of potent stress antigens89, and dermal γδ cells produce abun-
antigen-specific TRM cells at sites where the host is most dant IL-17 after C. albicans infection, application of
likely to encounter the pathogen in the future represents ­imiquimod or injection of IL-23 (refs99–101).
sound immunological planning. Two recent studies suggest an intriguing explanation
Latent HSV infection provides an illustrative model for the abundance of γδ T cells over αβ T cells in the
in which CD8+ TRM cells control active viral infection epidermis and dermis of young mice kept in SPF condi-
even when inflammation and effector cell recruit- tions. Mice infected with HSV developed HSV-specific
ment from the blood have abated81,82. Cutaneous HSV CD8+ T cells that assumed a dendritic morphology and
infection in mouse models results in the generation of displaced local DETCs68. Similarly, mice infected with
CD8+ TRM cells that remain millimetres from the site C. albicans developed a large population of CD4+
of infection, providing long-lasting site-specific immune TRM cells, which outnumbered dermal γδ cells and were
surveillance68,78. Protection against recrudescent HSV the predominant IL-17-producing cells on re-challenge76.
infection depends on HSV-specific TRM cells at the site In a non-SPF world replete with pathogens, such encoun-
of prior infection; these TRM cells engage virus-infected ters would likely happen repetitively over the life of
cells, are constrained to the epidermis and prolifer- the mouse. Indeed, the density of skin γδ cells in mice
ate in situ45. Furthermore, the cutaneous TRM cells are housed in pet stores or living in barns is diminished, and
maintained as a stable population after recall32,45. These αβ T cells are far more abundant, as are memory T cells

Nature Reviews | Immunology


Reviews

in general102. The immune systems of these mice more production, which is followed by production of IL-17
closely resemble those of adult humans, whereas those and IFNγ from dermal T cells116. A recent study has
of SPF mice resemble the immune systems of neonatal demonstrated that introduction of Staphylococcus epi-
humans102. One could speculate that skin γδ cells repre- dermidis to gnotobiotic mice restores the production
sent a primitive adaptive immune system that is replaced of IL-17A by T cells, indicating that S. epidermidis can
by αβ TRM cells over time as the mouse encounters more influence host defence as part of the skin commensal
and more antigenic pathogens through skin. microbiome. In this study, S. epidermis was the only
commensal species that increased the frequency of
CD1a and skin-specific T cell responses. Conventional CD8β+ T cells in the skin; these T cells reside in epi-
cutaneous T cell responses have been thought to require dermis and resemble TRM cells, enhance barrier immu-
T cell recognition of peptide antigens bound to MHC nity and limit pathogen invasion117. It is not known how
complexes on the cell surface of antigen-presenting many skin TRM cells are specific for antigens on commen-
cells. However, recent evidence has demonstrated that sal organisms and whether this putative responsiveness
T cells can respond to lipid antigens presented by CD1 is modified by the activity of skin Treg cells.
molecules103. Mice do not express CD1a, CD1b and
CD1c, whereas human skin Langerhans cells (LCs) Regulatory T cells in the skin
constitutively express CD1a at high levels104. Subsets of Barrier tissues also require mechanisms to regulate
human dermal DCs also express CD1a, albeit at lower and suppress inflammation 118–122. In human skin,
levels104. In contrast to the limited repertoire of invar- Treg cells have been described as a resident population
iant natural killer T (NKT) cells, the CD1a-specific and shown to interact with fibroblasts and LCs in situ
T cell repertoire contains diverse TCRs105,106. Many in the absence of inflammation13,15,54,79,123. In peripheral
CD1a-autoreactive T cells home to skin, where they pro- blood, most circulating Treg cells express skin-homing
duce IFNγ, IL-2 and IL-22 in response to CD1a–lipid markers, suggesting constitutive trafficking to skin124.
complexes on LCs107,108. Recent studies have suggested In mouse skin, a wave of Treg cells derived from the thymus
a role for CD1a-autoreactive T cells in mediating cuta- arises in early neonatal life in response to skin coloni-
neous allergic and inflammatory responses109–111. For zation with S. epidermidis, aiding in the establishment
example, in psoriatic skin lesions, there is an increased of immune tolerance to commensals97. In a model of
number of CD1a-reactive T cells associated with higher cutaneous inducible ovalbumin expression, a population
expression of mast cell-derived phospholipase A2 of Treg cells remain in the skin following the resolution of
(PLA2); this can convert non-antigenic phosphatidic skin inflammation and express low levels of CD25
acid-containing molecules into lysolipid neo-antigens and other markers associated with memory T cells125.
that can be accommodated by the groove of the CD1a However, inflammation-experienced Treg cells reverse
molecule109,110. Mice lack CD1a expression, and a trans- most of their activation-induced changes and lose their
genic mouse expressing the human CD1a transgene in suppressive ability over time126.
LCs showed exacerbated skin inflammation in response Advances in genomic technologies have provided
to imiquimod-induced dermatitis, which models cer- valuable tools to probe the diversity of tissue-resident
tain downstream events in psoriasis111. Furthermore, Treg cells. Single-cell RNA sequencing (scRNAseq) anal-
both bee venom-allergic individuals and house dust ysis of Treg cells from mouse colon and skin demonstrates
mite-allergic individuals have a higher frequency of a shared core transcriptional signature, suggesting that
CD1a-reactive T cells that release IFNγ, GM-CSF and there may be a shared general residency programme in
IL-13 in response to PLA2, likely derived from autol- barrier tissues127. Another recent study combined scR-
ogous substrates110. While CD1a-autoreactive T cells NAseq with TCR analysis of Treg cells from mouse colon
produce pro-inflammatory cytokines, the propensity and demonstrated that Treg cells sharing the same TCR
for IL-22 production by CD1a-autoreactive T cells in clonotype display similar transcriptional identities, sug-
healthy individuals suggests a possible immune home- gesting that shared antigenic specificity may drive these
ostasis function in skin109. There is also evidence that Treg cells to the same anatomical location and maintain
CD1 autoreactivity can serve an instructive or regulatory them there128. However, tissue-specific signals may shape
role via crosstalk with CD1-expressing DCs112. The com- the more nuanced transcriptional differences between
plete role of CD1a-reactive T cells in human skin disease tissue-specific Treg cells. Integrating scRNAseq with assay
remains to be elucidated. for transposase-accessible chromatin using sequencing
(ATACseq) to provide insight into chromatin accessibil-
The skin microbiota and T cell education. In recent ity of Treg cells isolated from the non-lymphoid tissues
years, the importance of commensal microorganisms in — visceral adipose tissue, colon and muscle — revealed
shaping the host immune system has become increas- that chromatin accessibility and gene expression often
ingly clear. Across the 1.8 m2 of the skin surface of the did not align, with a number of open chromatin regions
human body resides more than 1010 bacterial cells113. already accessible in the spleen129. It is tempting to specu-
Moreover, eukaryotic fungi and viruses are also abun- late that this mechanism may permit more rapid changes
dant114. The skin microbiome promotes both innate and in gene expression within tissues as Treg cells respond to
adaptive immune responses to limit pathogen invasion various tissue-specific stimuli.
and maintain homeostasis97,115. As part of their immune Recent work has also focused on the mechanisms by
modulatory role, skin commensals tune the function which Treg cells traffic to the skin. CCR4 has been a focus
of nearby T cells by stimulating increased DC IL-1α of study, as both circulating124 and cutaneous54 Treg cells

www.nature.com/nri
Reviews

Table 1 | Selected therapies that target pathogenic cutaneous T cells the epidermal growth factor receptor pathway, similar
to Treg cells involved in lung repair132.
Therapy mode of action Disease
Dupilumab Inhibits IL-4 and IL-13 Atopic dermatitis T cells in skin diseases
signalling The prevalence of T cell-mediated skin diseases and
Ixekizumab Neutralizes IL-17A Psoriasis their frequent responsiveness to skin-directed therapy
Secukinumab Neutralizes IL-17A Psoriasis have led to speculation that many or most of these dis-
eases involve TRM cells. A comprehensive discussion of
Ustekinumab Neutralizes p40 subunit Psoriasis
of IL-12 and IL-23 all T cell-mediated skin diseases is outside the scope
of this Review, but below we discuss some of the more
Tildrakizumab Neutralizes p19 subunit Psoriasis
of IL-23 common diseases, including psoriasis, atopic dermatitis,
alopecia areata and vitiligo.
Guselkumab Neutralizes p19 subunit Psoriasis
of IL-23
T cells in psoriasis. Psoriasis is a disease that afflicts
Adalimumab Neutralizes TNF • Psoriasis
2–4% of the world’s population. It is rarely fatal but can
• Hidradenitis suppurativa
• Pyoderma gangrenosum (off-label) be associated with crippling joint disease as well as a
pro-inflammatory metabolic syndrome that increases
Infliximab Neutralizes TNF • Psoriasis
• Hidradenitis suppurativa the risk of death from coronary artery disease, stroke
• Pyoderma gangrenosum (off-label) and adult-onset diabetes134,135. An immune aetiology for
Etanercept Inhibits TNF signalling • Psoriasis psoriasis had been suspected ever since the identification
• Hidradenitis suppurativa of HLA-Cw6 as a high-risk allele136,137. A seminal study
• Pyoderma gangrenosum (off-label) showed that denileukin diftitox (a drug comprising the
Tofacitinib JAK1 and JAK3 inhibitor • Psoriasis (off-label) ligand-binding portion of the IL-2R molecule fused to
• Alopecia areata (off-label) diphtheria toxin A subunit) cleared disease symptoms
• Vitiligo (off-label) in a significant number of patients with psoriasis, con-
• Atopic dermatitis (off-label) firming that T cells are necessary for clinical psoriasis138.
• Dermatomyositis (off-label)
Patients with psoriasis also responded impressively
Ruxolitinib JAK1 and JAK3 inhibitor • Psoriasis (off-label) to tumour necrosis factor (TNF) inhibitors, which
• Alopecia areata (off-label)
• Vitiligo (off-label) became the drug of choice for psoriasis for many years135
• Atopic dermatitis (off-label) (Table 1). Around the time that ustekinumab, an anti-
• Dermatomyositis (off-label) body directed at the shared p40 chain of IL-12 and IL-23,
The mechanistic dissection of the immunopathogenesis of inflammatory cutaneous disorders began to show impressive clinical effects in psoriasis, sev-
has resulted in highly targeted and effective therapies. As tissue-resident memory T (TRM) cell eral groups identified IL-17 as a key cytokine in psoriasis,
immunobiology becomes better understood, novel therapeutics that specifically eradicate
pathogenic TRM cells may lead to durable responses in a number of chronic, inflammatory skin and TH17 cells and IL-17-producing CD8+ T cells (Tc17)
diseases. JAK , Janus-activated kinase; TNF, tumour necrosis factor. cells were shown to be abundant in lesional skin139–141.
It is well known that IL-23 is critical for the expansion
express CCR4. In mice, CCR4-deficient Treg cells failed of IL-17-producing T cells, and recent clinical studies
to reconstitute the cutaneous Treg cell compartment130. using therapeutic antibodies against IL-17 or IL-23 (p19
Mouse Treg cells have been found to localize to hair fol- subunit) have shown the most dramatic improvement to
licles79,115, and in a mouse model of skin-specific hair date in psoriasis142–146 (Fig. 3). A number of studies have
follicle morphogenesis arrest, the skin Treg cell popula- shown that a single-nucleotide polymorphism in IL-23R
tion was reduced without affecting Treg cell populations is linked to human autoimmune disease147–151, raising the
in other tissues. Furthermore, the skin of neonatal SPF issue of whether IL-23R does something more than sim-
mice produces high amounts of CCL20, and its receptor, ply expand the number of TH17 cells. Regulatory network
CCR6, is enriched on cutaneous Treg cells. Accordingly, analysis suggests that IL-23 promotes the differentiation
adoptive transfer experiments demonstrated that CCR6- of pathogenic TH17 cells by inducing the phosphory­
deficient Treg cells were at a competitive disadvantage in lation of FOXO1 and unabated expression of IL-23R152.
reconstituting the skin T cell compartment115. CCL20 Thus, while TH17 cells can be expanded by IL-21, it is
mRNA expression is also increased in human fetal exposure to IL-23 that appears to be critical for evoking
skin explants exposed to cutaneous commensals and the pathogenic potential in TH17 cells. Transcriptomic
bacterial components115. analyses have elucidated transcriptional and regulatory
Very recently, Treg cells have been implicated in a networks that mediate TH17 cell differentiation and func-
number of non-immunological functions relevant tion152,153. Thus, the prediction would be that pharma-
to cutaneous biology. Cutaneous Treg cells have been cological inhibition of IL-23 would lead to even more
shown to regulate cutaneous wound healing and, in durable responses than inhibition of IL-17 and may not
some models, to modulate hair follicle stem cells131–133. result in the paradoxical worsening of inflammatory
Accordingly, skin Treg cells accumulate in large numbers bowel disease seen with IL-17 inhibitors154,155.
at the affected site soon after a wound injury in skin. Psoriasis has a predilection for always return-
These Treg cells have an activated phenotype and limit ing in the same anatomical location, and a recent
IFNγ production by T cells and inflammatory macro­ study showed that healed psoriasis lesions cleared by
phages in wounds133. The Treg cells involved in cuta­ anti-TNF therapy contained TRM cell clones, several of
neous wound healing were shown to be dependent on which shared the same amino acid sequence in the third

Nature Reviews | Immunology


Reviews

Epidermis

Stimulus (trauma, infections,


medication or release of
cytokines and/or IL-1α)

TNF Tc17 cell


(TRM cell)
Autoantigens
LL-37/cathelicidin, Resident
ADAMTSL5, TH17 cell
PLA2G4D
IL-6, IL-17,
IL-23, IL-21, Epidermal remodelling
TGFβ IL-22 and/or keratinocyte
Dendritic cell
release of proinflammatory
cytokines

VEGF,
T17 cell FGF

Pro-inflammatory Angiogenesis
cycle
IL-23,
IL-6, T17 cell
TGFβ
Neutrophil
Secondary lymphoid tissue recruitment

Fig. 3 | T cells in the pathogenesis of psoriasis. After an initial stimulus that results in the activation of the innate immune
system and release of putative auto-antigens, activated dendritic cells drive the activation and expansion of IL-17+
tissue-resident memory (TRM) cells (T17 cells), which include both IL-17-producing CD8+ T cells (Tc17 cells) and T helper 17
(TH17) cells. IL-23 is critical for the expansion and the differentiation of pathogenic T17 cells. The development of
pathogenic T17 cells leads to the release of a pro-inflammatory cytokine milieu resulting in the key features of psoriasis
including neutrophil microabscesses, keratinocyte proliferation, further lymphocytic infiltration and angiogenesis. IL-22,
produced by either Tc17 cells or TH22 cells, also drives epidermal proliferation. TNF, tumour necrosis factor.

complementarity-determining region (CDR3) of their on clinical features, as there are no precise consensus
TCR156. Because anti-TNF treatment will suppress the laboratory or histological findings158,159. Eczematous,
activity of TRM cells but cannot dislodge them from cutaneous lesions in a characteristic distribution, which
the tissue, it is likely that these antigen-specific TRM cells changes depending on age of onset, and severe pruri-
that remain at the same site will be activated in the tus are hallmarks of disease158,159. Moreover, AD is often
future. However, the antigen specificity of these CD8+ αβ associated with different atopic and allergic comor-
T cells is unknown. Although mouse models of psoriasis, bidities, including food allergies, allergic rhinitis and
whether mediated by topical imiquimod application or asthma, all disorders associated with overdeveloped
IL-23 injection, exclusively involve γδ T cells produc- type 2 immunity158,159. Though the precise pathogenesis
ing IL-17, these cells appear to be a small minority in of AD continues to be examined, two major mechanisms
human skin, whether normal or psoriatic. As shown by have emerged: defects in the skin barrier and immune
high-throughput sequencing of TCRs, γδ cells make dysregulation160,161. AD pathogenesis likely results from
up <3% of T cells in psoriatic skin and are not observed an interplay of these two mechanisms, but immune dys-
at all in healed psoriatic lesions156. The elucidation of the function appears to play an outsized role in the develop-
immune pathophysiology of psoriasis remains the most ment of AD. Cutaneous barrier defects resulting from
complete and comprehensive body of work on a human filaggrin mutations are reviewed elsewhere162.
autoimmune disease. Non-lesional skin from patients with AD demon-
strates evidence of subclinical inflammation with a
T cells in atopic dermatitis. Atopic dermatitis (AD) is pro-inflammatory cytokine milieu and increased num-
the most common chronic inflammatory skin disease, bers of TH2 cells, TH22 cells and, to a lesser degree,
with the lifetime prevalence reaching 10–20% in devel- TH17 cells163. However, immune signatures from the
oped countries157. The diagnosis of AD relies heavily lesional skin of adult and paediatric patients with AD

www.nature.com/nri
Reviews

suggest that there may be subtle differences in the dis- the body (alopecia universalis) can occur174. The exact
ease process, which may be age related163. Children have pathogenesis of alopecia areata is an area of active inves-
strong activation of TH2 cell-type and TH22 cell-type tigation, but evidence suggests that genetic and environ-
cytokines; however, paediatric skin also has levels of mental factors cause an autoimmune reaction to the hair
IL-9, IL-33 and TH17 cell-associated cytokines that are follicles175. A genome-wide association study implicated
higher than those in adults163. Filaggrin mutations can HLA-DR and several genes involved in the regulation of
predispose to the development of AD, and epidermal Treg cells and cytotoxic T cells in disease susceptibility176.
barrier disruptions and skin inflammation are mutually A recent meta-analysis confirmed that HLA-DR is the
reinforcing processes, as perturbations of the epidermal largest risk factor for alopecia areata. These MHC class II
barrier intrinsically stimulate inflammation by activat- genes are highly linked to the functions of CD4+ T cells,
ing keratinocytes to release disease-associated cytokines which are important effector cells in alopecia areata177.
and chemokines164,165. In humans with AD, peripheral Experimental evidence also points to the importance
blood CCR8+CD4+ T cells are enriched for IL-5 expres- of CD8+ T cells in mediating the alopecia areata disease
sion, whereas CCR4+CD4+ T cells are enriched for IL-4 process. The hair follicle is an immune-privileged site,
(ref. 166) . Furthermore, thymic stromal lymphopoie- with low levels of MHC expression178. A proposed model
tin (TSLP) is expressed by epithelial cells, especially of alopecia areata pathogenesis suggests that a break in
keratinocytes, from patients with AD167. TSLP acti- immune privilege results in a subsequent assault on the
vates CD11c+ DCs and induces the production of TH2 hair follicle at the level of the bulb by oligoclonal and
cell-attracting chemokines. TSLP-activated DCs polarize autoreactive CD8+ T cells178. The peribulbar lymphocytic
naive CD4+ T cells to produce IL-4, IL-5, IL-13 and TNF infiltrate induces hair follicle keratinocytes to undergo
while downregulating IL-10 and IFNγ167. TSLP appears apoptosis that results in inhibition of cell division within
to have a dual role in that it directly communicates the hair matrix and of hair shaft production179. In one
with somatosensory nerves and induces TH2 cell-type study in which scalp explants from patients with alo-
cytokines such as IL-13 and IL-31, which stimulate nerve pecia areata were transplanted into severe combined
fibres directly and upregulate the release of cellular pru- immunodeficient (SCID) mice and injected with autolo-
ritogens. The resulting neurogenic inflammation com- gous T cells isolated from involved scalp, T cells that had
pletes the vicious cycle by promoting TH2 cell responses, been cultured in the presence (but not the absence) of
keratinocyte proliferation and epidermal thickening168. hair follicle homogenate induced changes reminiscent
Recently available and highly successful AD treat- of alopecia areata, including hair loss and perifollicular
ments highlight the critical importance of type 2 immune infiltrates of T cells, along with HLA-DR and ICAM1
responses to disease pathogenesis and the pathogenic expression of the follicular epithelium180. Furthermore,
role of epidermal dysfunction in disease pathogenesis. cytotoxic CD8+NKG2D+ T cells are both necessary and
Dupilumab is a monoclonal antibody that targets the sufficient for the induction of alopecia areata in a mouse
IL-4Rα chain, which is shared by the receptors for IL-4 model of disease181. Transcriptional profiling of mouse
and IL-13 (Table 1). Clinical trials have demonstrated its and human alopecia areata skin revealed gene expres-
remarkable efficacy in the treatment of AD169. If psoriasis sion signatures indicative of cytotoxic T cell infiltration,
can be described as a disease of the IL-17–IL-23 axis, AD an IFNγ response and upregulation of several γ-chain
appears to be a disease of the IL-4–IL-13 axis. The degree cytokines known to promote the activation and survival
to which this is true for atopic diseases of other tissues of IFNγ-producing CD8+NKG2D+ effector T cells181.
(such as asthma) remains to be elucidated. Therapeutic Therapeutic targeting of Janus-activated kinases (JAKs,
targeting of the downstream results of epithelial damage which are key signalling molecules downstream of the
may provide additional insights into AD pathogenesis IFNγ receptor) has established the central role of IFNγ
and novel treatments. IL-33 is produced by a number of in alopecia areata disease pathogenesis. Several reports
cell types including keratinocytes and, along with TSLP, have demonstrated hair regrowth in patients with alope-
is a potent stimulator of allergic type 2 inflammation170. cia areata and in mouse models upon treatment with JAK
This makes IL-33 a particularly attractive target in AD inhibitors181,182 (Table 1). Multiple clinical trials involving
and, indeed, this is an area of active investigation. IL-22 systemic and topical JAK inhibitors are underway183.
also plays an important role in maintaining the epithe-
lial barrier171, and clinical trials using an antibody to T cells in vitiligo. Vitiligo is a cutaneous autoimmune
target IL-22 in AD have shown promise172, suggesting disorder that results in white, depigmented patches of
that IL-22 plays a pathogenic role in AD. Indeed, novel skin owing to the destruction of melanocytes by the
treatments that target type 2 immune responses and bar- immune system184. A better understanding of vitiligo
rier dysfunction may improve the array of therapeutic disease pathogenesis has led to new treatment strate-
options for patients with AD. gies. Both immune dysfunction and intrinsic melano-
cyte abnormalities are proposed mechanisms of disease
T cells in alopecia areata. Alopecia areata is a cutane- initiation185. Accordingly, CD8+ T cells are both neces-
ous disorder that results in non-scarring hair loss with sary and sufficient to mediate melanocyte destruction
a cumulative lifetime incidence of 2%173. The diagno- in human subjects and have been a focus of active inves-
sis of alopecia areata is made clinically, and the disease tigation186. Similarly, melanocytes from patients with
typically manifests with sharply demarcated, annular vitiligo grow poorly ex vivo and are more susceptible
patches of hair loss without atrophy. Less commonly, loss to oxidative stress187. The role of oxidative stress in the
of all scalp hair (alopecia totalis) or all terminal hair on pathogenesis of vitiligo is reviewed elsewhere188.

Nature Reviews | Immunology


Reviews

Recent evidence has identified CD8+ T cells as key very recently permitted an explosion of novel ther-
drivers of vitiligo disease progression186. Gene expression apies for T cell-mediated skin disorders. A derma-
analysis of human lesional skin revealed upregulation of tologist treating moderate to severe psoriasis may
IFNγ and IFNγ-dependent genes, including those encod- choose between highly targeted biologics directed
ing CXCR3 and its ligands, CXCL9, CXCL10 and CXCL11 at TNF, IL-17 or IL-23, options that were unthink-
(ref.189). These findings were consistent with functional able in the 20th century. Therapeutic antibodies to
studies in mouse models of vitiligo190. Disruption of the the receptor for IL-4 and IL-13 have revolutionized the
IFNγ–chemokine signalling axis prevents T cell recruit- treatment of AD and demonstrated that T cells are
ment to the skin, subsequent melanocyte destruction central to this disorder. As the immunopathological
and pigment loss. Furthermore, blockade of this signal- beginnings of alopecia areata and vitiligo are mech-
ling axis not only prevented vitiligo disease in mice but anistically dissected, JAK inhibitors targeting T H1
also reversed established disease190,191. These findings led cell cytokines have emerged as promising therapies197
to therapeutic targeting of the IFNγ signalling pathway (Table 1) . There are a host of other T cell-mediated
clinically with JAK inhibitors. Patients with vitiligo that skin disorders, which, when studied mechanistically,
received either of the JAK inhibitors tofacitinib or ruxol- may result in the development of new therapies or a
itinib rapidly repigmented their skin. A patient treated novel use for an existing drug. Many other therapeu-
with ruxolitinib was found to have an increased serum tic antibodies directed against cytokines and chemo­
level of CXCL10, which rapidly decreased after treatment kines are either being developed or already in clinical
with the JAK inhibitor192–194 (Table 1). This observation trials143,198,199. Furthermore, as discussed above, TRM cells
was consistent with the hypothesis that the mechanism of have been demonstrated as fundamentally important
improvement was through inhibition of IFNγ signalling in normal and diseased skin2,3 and are the likely targets
and resulting loss of chemokine production. of these drugs. Looking backwards, dermatologists
Vitiligo lesions frequently recur at previous sites have successfully treated T cell-mediated skin disor-
upon treatment cessation, suggesting a potential role for ders with skin-directed therapies (including topical
TRM cells. A recent report demonstrated that TRM cells medicines and ultraviolet light) for more than 100
are highly enriched in the skin of patients with vitiligo years, unaware that their target was the then undis-
and show cytotoxic effector functions when exposed to covered pathogenic skin TRM cells. The next frontier
inflammatory cytokines195. Furthermore, another group may involve dislodging pathogenic TRM cells rather
found that TRM cells are present in vitiligo lesions in a than simply episodically suppressing their function,
mouse model of melanoma and vitiligo and might be as all current therapies do. If pathogenic TRM cells can
responsible in part for mediating protection against be dislodged while preserving at least some protective
recurrence of melanoma 196. Antibody blockade of TRM cell function, durable remissions may be possi-
CD122 (a component of the IL-15R) in a mouse model ble2,3,200. Finally, it is important to emphasize that the
of vitiligo results in the depletion of pathogenic TRM cells scientific study of immune-mediated skin diseases can
in lesional skin and reversal of disease69. Future stud- be more readily accomplished than that of any other
ies might determine that targeting TRM cells in patients human organ, largely owing to its accessibility. While
with vitiligo could be an effective treatment strategy and immune-mediated diseases of other organs will have
could be durable, even after discontinuing treatment. unique features, there is a great deal of common mech-
anism, and it is inevitable that the study of skin immu-
Conclusions and future directions nology and immunopathology will inform all human
Ultimately, the study of cutaneous T cells has as its immune-mediated disorders.
goal the improvement of human health. Our new
knowledge of the basic immunobiology of skin has Published online xx xx xxxx

1. Sathaliyawala, T. et al. Distribution and 9. Kupper, T. S. The activated keratinocyte: a model for 16. Gaide, O. et al. Common clonal origin of central and
compartmentalization of human circulating and inducible cytokine production by non-bone resident memory T cells following skin immunization.
tissue-resident memory T cell subsets. Immunity 38, marrow-derived cells in cutaneous inflammatory and Nat. Med. 21, 647–653 (2015).
187–197 (2013). immune responses. J. Invest. Dermatol. 94, 17. Jiang, X. et al. Skin infection generates non-migratory
2. Park, C. O. & Kupper, T. S. The emerging role of 146S–150S (1990). memory CD8+ T(RM) cells providing global skin
resident memory T cells in protective immunity 10. Lande, R. et al. Plasmacytoid dendritic cells sense immunity. Nature 483, 227–231 (2012).
and inflammatory disease. Nat. Med. 21, 688–697 self-DNA coupled with antimicrobial peptide. Nature 18. Ahmed, R., Bevan, M. J., Reiner, S. L. & Fearon, D. T.
(2015). 449, 564–569 (2007). The precursors of memory: models and controversies.
3. Clark, R. A. Resident memory T cells in human 11. Kashem, S. W., Haniffa, M. & Kaplan, D. H. Nat. Rev. Immunol. 9, 662–668 (2009).
health and disease. Sci. Transl Med. 7, 269rv261 Antigen-presenting cells in the skin. Annu. Rev. 19. Chang, J. T. et al. Asymmetric T lymphocyte division in
(2015). Immunol. 35, 469–499 (2017). the initiation of adaptive immune responses. Science
4. Pasparakis, M., Haase, I. & Nestle, F. O. Mechanisms 12. Pasparakis, M. et al. TNF-mediated inflammatory skin 315, 1687–1691 (2007).
regulating skin immunity and inflammation. Nat. Rev. disease in mice with epidermis-specific deletion of 20. Youngblood, B. et al. Effector CD8 T cells
Immunol. 14, 289–301 (2014). IKK2. Nature 417, 861–866 (2002). dedifferentiate into long-lived memory cells. Nature
5. Kupper, T. S. & Fuhlbrigge, R. C. Immune surveillance 13. Clark, R. A. et al. The vast majority of CLA+ T cells 552, 404–409 (2017).
in the skin: mechanisms and clinical consequences. are resident in normal skin. J. Immunol. 176, 21. Akondy, R. S. et al. Origin and differentiation of
Nat. Rev. Immunol. 4, 211–222 (2004). 4431–4439 (2006). human memory CD8 T cells after vaccination. Nature
6. Elias, P. M. The skin barrier as an innate immune 14. Mueller, S. N., Gebhardt, T., Carbone, F. R. & 552, 362–367 (2017).
element. Semin. Immunopathol. 29, 3–14 (2007). Heath, W. R. Memory T cell subsets, migration 22. Carbone, F. R. Tissue-resident memory T cells and
7. Burgeson, R. E. & Christiano, A. M. The dermal- patterns, and tissue residence. Annu. Rev. Immunol. fixed immune surveillance in nonlymphoid organs.
epidermal junction. Curr. Opin. Cell Biol. 9, 651–658 31, 137–161 (2013). J. Immunol. 195, 17–22 (2015).
(1997). 15. Watanabe, R. et al. Human skin is protected by four 23. Mackay, C. R. et al. Tissue-specific migration
8. Robert, C. & Kupper, T. S. Inflammatory skin diseases, functionally and phenotypically discrete populations pathways by phenotypically distinct subpopulations
T cells, and immune surveillance. N. Engl. J. Med. of resident and recirculating memory T cells. of memory T cells. Eur. J. Immunol. 22, 887–895
341, 1817–1828 (1999). Sci. Transl Med. 7, 279ra239 (2015). (1992).

www.nature.com/nri
Reviews

24. Mackay, C. R., Andrew, D. P., Briskin, M., Ringler, D. J. phenotype and migratory pattern. J. Immunol. 190, during viral infection. J. Exp. Med. 213, 951–966
& Butcher, E. C. Phenotype, and migration properties 970–976 (2013). (2016).
of three major subsets of tissue homing T cells in 49. Hogan, R. J. et al. Protection from respiratory virus 73. Schenkel, J. M., Fraser, K. A., Vezys, V. & Masopust, D.
sheep. Eur. J. Immunol. 26, 2433–2439 (1996). infections can be mediated by antigen-specific CD4+ Sensing and alarm function of resident memory CD8+
25. Masopust, D., Vezys, V., Marzo, A. L. & Lefrancois, L. T cells that persist in the lungs. J. Exp. Med. 193, T cells. Nat. Immunol. 14, 509–513 (2013).
Preferential localization of effector memory cells in 981–986 (2001). 74. Ariotti, S. et al. T cell memory. Skin-resident memory
nonlymphoid tissue. Science 291, 2413–2417 50. Reinhardt, R. L., Khoruts, A., Merica, R., Zell, T. CD8+ T cells trigger a state of tissue-wide pathogen
(2001). & Jenkins, M. K. Visualizing the generation of memory alert. Science 346, 101–105 (2014).
26. Liu, L., Fuhlbrigge, R. C., Karibian, K., Tian, T. CD4 T cells in the whole body. Nature 410, 101–105 75. Casey, K. A. et al. Antigen-independent differentiation
& Kupper, T. S. Dynamic programming of CD8+ T cell (2001). and maintenance of effector-like resident memory
trafficking after live viral immunization. Immunity 25, 51. Marshall, D. R. et al. Measuring the diaspora for T cells in tissues. J. Immunol. 188, 4866–4875
511–520 (2006). virus-specific CD8+ T cells. Proc. Natl Acad. Sci. USA (2012).
27. Mora, J. R., Iwata, M. & von Andrian, U. H. Vitamin 98, 6313–6318 (2001). 76. Park, C. O. et al. Staged development of long-lived
effects on the immune system: vitamins A and D take 52. Teraki, Y. & Shiohara, T. IFN-gamma-producing effector T cell receptor alphabeta TH17 resident memory
centre stage. Nat. Rev. Immunol. 8, 685–698 (2008). CD8+ T cells and IL-10-producing regulatory CD4+ T cell population to Candida albicans after skin
28. Sigmundsdottir, H. & Butcher, E. C. Environmental T cells in fixed drug eruption. J. Allergy Clin. Immunol. infection. J. Allergy Clin. Immunol. 142, 647–662
cues, dendritic cells and the programming of 112, 609–615 (2003). (2017).
tissue-selective lymphocyte trafficking. Nat. Immunol. 53. Boyman, O. et al. Spontaneous development of 77. Glennie, N. D. et al. Skin-resident memory CD4+ T cells
9, 981–987 (2008). psoriasis in a new animal model shows an essential enhance protection against Leishmania major
29. Sigmundsdottir, H. et al. DCs metabolize sunlight- role for resident T cells and tumor necrosis infection. J. Exp. Med. 212, 1405–1414 (2015).
induced vitamin D3 to ‘program’ T cell attraction to factor-alpha. J. Exp. Med. 199, 731–736 (2004). 78. Gebhardt, T. et al. Different patterns of peripheral
the epidermal chemokine CCL27. Nat. Immunol. 8, 54. Clark, R. A. & Kupper, T. S. IL-15 and dermal migration by memory CD4+ and CD8+ T cells. Nature
285–293 (2007). fibroblasts induce proliferation of natural regulatory 477, 216–219 (2011).
30. McCully, M. L. et al. Epidermis instructs skin homing T cells isolated from human skin. Blood 109, 79. Sanchez Rodriguez, R. et al. Memory regulatory T cells
receptor expression in human T cells. Blood 120, 194–202 (2007). reside in human skin. J. Clin. Invest. 124, 1027–1036
4591–4598 (2012). 55. Thome, J. J. & Farber, D. L. Emerging concepts (2014).
31. Yamanaka, K. et al. Vitamins A and D are potent in tissue-resident T cells: lessons from humans. 80. Collins, N. et al. Skin CD4+ memory T cells exhibit
inhibitors of cutaneous lymphocyte-associated antigen Trends Immunol. 36, 428–435 (2015). combined cluster-mediated retention and equilibration
expression. J. Allergy Clin. Immunol. 121, 148–157 56. Schenkel, J. M. et al. T cell memory. Resident with the circulation. Nat. Commun. 7, 11514 (2016).
(2008). memory CD8 T cells trigger protective innate and 81. Himmelein, S. et al. Circulating herpes simplex type 1
32. Mackay, L. K. et al. The developmental pathway for adaptive immune responses. Science 346, 98–101 (HSV-1)-specific CD8+ T cells do not access HSV-1
CD103+CD8+ tissue-resident memory T cells of skin. (2014). latently infected trigeminal ganglia. Herpesviridae 2,
Nat. Immunol. 14, 1294–1301 (2013). 57. Salerno, E. P., Olson, W. C., McSkimming, C., 5 (2011).
33. Bergsbaken, T. & Bevan, M. J. Proinflammatory Shea, S. & Slingluff, C. L. Jr. T cells in the human 82. Wakim, L. M., Jones, C. M., Gebhardt, T., Preston, C. M.
microenvironments within the intestine regulate the metastatic melanoma microenvironment express & Carbone, F. R. CD8+ T cell attenuation of cutaneous
differentiation of tissue-resident CD8+ T cells site-specific homing receptors and retention integrins. herpes simplex virus infection reduces the average
responding to infection. Nat. Immunol. 16, 406–414 Int. J. Cancer 134, 563–574 (2014). viral copy number of the ensuing latent infection.
(2015). 58. Ganesan, A. P. et al. Tissue-resident memory features Immunol. Cell Biol. 86, 666–675 (2008).
34. Harris, T. H. et al. Generalized Levy walks and the role are linked to the magnitude of cytotoxic T cell 83. Zhu, J. et al. Virus-specific CD8+ T cells accumulate
of chemokines in migration of effector CD8+ T cells. responses in human lung cancer. Nat. Immunol. 18, near sensory nerve endings in genital skin during
Nature 486, 545–548 (2012). 940–950 (2017). subclinical HSV-2 reactivation. J. Exp. Med. 204,
35. Nakanishi, Y., Lu, B., Gerard, C. & Iwasaki, A. CD8+ T 59. Djenidi, F. et al. CD8+ CD103+ tumor-infiltrating 595–603 (2007).
lymphocyte mobilization to virus-infected tissue lymphocytes are tumor-specific tissue-resident 84. Zhu, J. et al. Immune surveillance by CD8alphaalpha+
requires CD4+ T cell help. Nature 462, 510–513 memory T cells and a prognostic factor for survival in skin-resident T cells in human herpes virus infection.
(2009). lung cancer patients. J. Immunol. 194, 3475–3486 Nature 497, 494–497 (2013).
36. Hickman, H. D. et al. CXCR3 chemokine receptor (2015). 85. Zhu, J. et al. Persistence of HIV-1 receptor-positive
enables local CD8+ T cell migration for the destruction 60. Li, J., Olshansky, M., Carbone, F. R. & Ma, J. Z. cells after HSV-2 reactivation is a potential mechanism
of virus-infected cells. Immunity 42, 524–537 (2015). Transcriptional analysis of T cells resident in human for increased HIV-1 acquisition. Nat. Med. 15,
37. Laidlaw, B. J. et al. CD4+ T cell help guides formation skin. PLOS ONE 11, e0148351 (2016). 886–892 (2009).
of CD103+ lung-resident memory CD8+ T cells during 61. Milner, J. J. et al. Runx3 programs CD8+ T cell 86. Mestas, J. & Hughes, C. C. Of mice and not men:
influenza viral infection. Immunity 41, 633–645 residency in non-lymphoid tissues and tumours. differences between mouse and human immunology.
(2014). Nature 552, 253–257 (2017). J. Immunol. 172, 2731–2738 (2004).
38. Liu, L. et al. Epidermal injury and infection during 62. Mackay, L. K. et al. T-Box transcription factors 87. Thome, J. J. et al. Spatial map of human T cell
poxvirus immunization is crucial for the generation of combine with the cytokines TGF-beta and IL-15 to compartmentalization and maintenance over decades
highly protective T cell-mediated immunity. Nat. Med. control tissue-resident memory T cell fate. Immunity of life. Cell 159, 814–828 (2014).
16, 224–227 (2010). 43, 1101–1111 (2015). 88. Kumar, B. V., Connors, T. J. & Farber, D. L. Human
39. Pan, Y. et al. Survival of tissue-resident memory 63. Cyster, J. G. & Schwab, S. R. Sphingosine-1-phosphate T cell development, localization, and function
T cells requires exogenous lipid uptake and and lymphocyte egress from lymphoid organs. throughout life. Immunity 48, 202–213 (2018).
metabolism. Nature 543, 252–256 (2017). Annu. Rev. Immunol. 30, 69–94 (2012). 89. MacLeod, A. S. et al. Dendritic epidermal T cells
40. Mackay, L. K. et al. Hobit and Blimp1 instruct a 64. Steinert, E. M. et al. Quantifying memory CD8 T cells regulate skin antimicrobial barrier function.
universal transcriptional program of tissue residency reveals regionalization of immunosurveillance. Cell J. Clin. Invest. 123, 4364–4374 (2013).
in lymphocytes. Science 352, 459–463 (2016). 161, 737–749 (2015). 90. Jameson, J. M., Cauvi, G., Witherden, D. A. &
41. Sallusto, F., Lenig, D., Forster, R., Lipp, M. 65. Park, S. L., Mackay, L. K. & Gebhardt, T. Distinct Havran, W. L. A keratinocyte-responsive gamma delta
& Lanzavecchia, A. Two subsets of memory T recirculation potential of CD69+ CD103− and CD103+ TCR is necessary for dendritic epidermal T cell
lymphocytes with distinct homing potentials and thymic memory CD8+ T cells. Immunol. Cell Biol. 94, activation by damaged keratinocytes and maintenance
effector functions. Nature 401, 708–712 (1999). 975–980 (2016). in the epidermis. J. Immunol. 172, 3573–3579
42. Campbell, J. J., Clark, R. A., Watanabe, R. & 66. Kumar, B. V. et al. Human tissue-resident memory (2004).
Kupper, T. S. Sezary syndrome and mycosis fungoides T cells are defined by core transcriptional and 91. Jiang, X., Campbell, J. J. & Kupper, T. S. Embryonic
arise from distinct T cell subsets: a biologic rationale functional signatures in lymphoid and mucosal sites. trafficking of gammadelta T cells to skin is dependent
for their distinct clinical behaviors. Blood 116, Cell Rep. 20, 2921–2934 (2017). on E/P selectin ligands and CCR4. Proc. Natl Acad.
767–771 (2010). 67. Borges da Silva, H. et al. The purinergic receptor Sci. USA 107, 7443–7448 (2010).
43. Campbell, J. J. et al. CCR7 expression and memory P2RX7 directs metabolic fitness of long-lived memory 92. Toulon, A. et al. A role for human skin-resident T cells
T cell diversity in humans. J. Immunol. 166, 877–884 CD8+ T cells. Nature 559, 264–268 (2018). in wound healing. J. Exp. Med. 206, 743–750
(2001). 68. Zaid, A. et al. Persistence of skin-resident memory (2009).
44. Gehad, A. et al. A primary role for human central T cells within an epidermal niche. Proc. Natl Acad. 93. Jameson, J. et al. A role for skin gammadelta T cells in
memory cells in tissue immunosurveillance. Blood Adv. Sci. USA 111, 5307–5312 (2014). wound repair. Science 296, 747–749 (2002).
2, 292–298 (2018). 69. Richmond, J. M. et al. Antibody blockade of IL-15 94. Adams, E. J., Gu, S. & Luoma, A. M. Human gamma
45. Park, S. L. et al. Local proliferation maintains a signaling has the potential to durably reverse vitiligo. delta T cells: evolution and ligand recognition. Cell.
stable pool of tissue-resident memory T cells after Sci. Transl Med. 10, eaam7710 (2018). Immunol. 296, 31–40 (2015).
antiviral recall responses. Nat. Immunol. 19, 70. Van Braeckel-Budimir, N., Varga, S. M., Badovinac, V. P. 95. Cruz, M. S., Diamond, A., Russell, A. & Jameson, J. M.
183–191 (2018). & Harty, J. T. Repeated antigen exposure extends the Human alphabeta and gammadelta T cells in skin
46. Beura, L. K. et al. Intravital mucosal imaging of CD8+ durability of influenza-specific lung-resident memory immunity and disease. Front. Immunol. 9, 1304
resident memory T cells shows tissue-autonomous CD8+ T cells and heterosubtypic immunity. Cell Rep. (2018).
recall responses that amplify secondary memory. Nat. 24, 3374–3382 (2018). 96. Jiang, X. et al. Dermal gammadelta T cells do not
Immunol. 19, 173–182 (2018). 71. Masopust, D., Vezys, V., Wherry, E. J., Barber, D. L. freely re-circulate out of skin and produce IL-17 to
47. Clark, R. A. et al. Skin effector memory T cells do not & Ahmed, R. Cutting edge: gut microenvironment promote neutrophil infiltration during primary contact
recirculate and provide immune protection in promotes differentiation of a unique memory CD8 hypersensitivity. PLOS ONE 12, e0169397 (2017).
alemtuzumab-treated CTCL patients. Sci. Transl Med. T cell population. J. Immunol. 176, 2079–2083 97. Scharschmidt, T. C. et al. A wave of regulatory T cells
4, 117ra117 (2012). (2006). into neonatal skin mediates tolerance to commensal
48. Bromley, S. K., Yan, S., Tomura, M., Kanagawa, O. 72. Khan, T. N., Mooster, J. L., Kilgore, A. M., Osborn, J. F. microbes. Immunity 43, 1011–1021 (2015).
& Luster, A. D. Recirculating memory T cells are a & Nolz, J. C. Local antigen in nonlymphoid tissue 98. Strid, J. et al. Acute upregulation of an NKG2D ligand
unique subset of CD4+ T cells with a distinct promotes resident memory CD8+ T cell formation promotes rapid reorganization of a local immune

Nature Reviews | Immunology


Reviews

compartment with pleiotropic effects on 127. Miragaia, R. J. et al. Single cell transcriptomics of double-blind placebo-controlled trial. Gut 61,
carcinogenesis. Nat. Immunol. 9, 146–154 (2008). regulatory T cells reveals trajectories of tissue 1693–1700 (2012).
99. Kashem, S. W. et al. Nociceptive sensory fibers drive adaptation. Immunity 50, 493–504 (2017). 155. Fobelo Lozano, M. J., Serrano Gimenez, R. &
interleukin-23 production from CD301b+ dermal 128. Zemmour, D. et al. Single-cell gene expression reveals Castro Fernandez, M. Emergence of inflammatory
dendritic cells and drive protective cutaneous a landscape of regulatory T cell phenotypes shaped by bowel disease during treatment with secukinumab.
immunity. Immunity 43, 515–526 (2015). the TCR. Nat. Immunol. 19, 291–301 (2018). J. Crohns Colitis 12, 1131–1133 (2018).
100. Cai, Y. et al. Pivotal role of dermal IL-17-producing 129. DiSpirito, J. R. et al. Molecular diversification of 156. Matos, T. R. et al. Clinically resolved psoriatic lesions
gammadelta T cells in skin inflammation. Immunity regulatory T cells in nonlymphoid tissues. Sci. Immunol. contain psoriasis-specific IL-17-producing αβ T cell
35, 596–610 (2011). 3, eaat5861 (2018). clones. J. Clin. Invest. 127, 4031–4041 (2017).
101. Komori, H. K. et al. Cutting edge: dendritic epidermal 130. Sather, B. D. et al. Altering the distribution of Foxp3+ 157. Deckers, I. A. et al. Investigating international time
gammadelta T cell ligands are rapidly and locally regulatory T cells results in tissue-specific trends in the incidence and prevalence of atopic
expressed by keratinocytes following cutaneous inflammatory disease. J. Exp. Med. 204, 1335–1347 eczema 1990–2010: a systematic review of
wounding. J. Immunol. 188, 2972–2976 (2012). (2007). epidemiological studies. PLOS ONE 7, e39803 (2012).
102. Beura, L. K. et al. Normalizing the environment 131. Ali, N. et al. Regulatory T cells in skin facilitate 158. Guttman-Yassky, E., Nograles, K. E. & Krueger, J. G.
recapitulates adult human immune traits in laboratory epithelial stem cell differentiation. Cell 169, Contrasting pathogenesis of atopic dermatitis and
mice. Nature 532, 512–516 (2016). 1119–1129 (2017). psoriasis — part II: immune cell subsets and
103. Van Rhijn, I., Godfrey, D. I., Rossjohn, J. & Moody, D. B. 132. Arpaia, N. et al. A distinct function of regulatory therapeutic concepts. J. Allergy Clin. Immunol. 127,
Lipid and small-molecule display by CD1 and MR1. T cells in tissue protection. Cell 162, 1078–1089 1420–1432 (2011).
Nat. Rev. Immunol. 15, 643–654 (2015). (2015). 159. Guttman-Yassky, E., Nograles, K. E. & Krueger, J. G.
104. Dougan, S. K., Kaser, A. & Blumberg, R. S. CD1 133. Nosbaum, A. et al. Cutting edge: regulatory T cells Contrasting pathogenesis of atopic dermatitis and
expression on antigen-presenting cells. Curr. Top. facilitate cutaneous wound healing. J. Immunol. 196, psoriasis — part I: clinical and pathologic concepts.
Microbiol. Immunol. 314, 113–141 (2007). 2010–2014 (2016). J. Allergy Clin. Immunol. 127, 1110–1118 (2011).
105. Mori, L., Lepore, M. & De Libero, G. The immunology 134. Nestle, F. O., Kaplan, D. H. & Barker, J. Psoriasis. 160. Brunner, P. M., Guttman-Yassky, E. & Leung, D. Y.
of CD1- and MR1-restricted T cells. Annu. Rev. N. Engl. J. Med. 361, 496–509 (2009). The immunology of atopic dermatitis and its
Immunol. 34, 479–510 (2016). 135. Lowes, M. A., Bowcock, A. M. & Krueger, J. G. reversibility with broad-spectrum and targeted
106. Godfrey, D. I., Stankovic, S. & Baxter, A. G. Raising the Pathogenesis and therapy of psoriasis. Nature 445, therapies. J. Allergy Clin. Immunol. 139, S65–S76
NKT cell family. Nat. Immunol. 11, 197–206 (2010). 866–873 (2007). (2017).
107. de Jong, A. et al. CD1a-autoreactive T cells recognize 136. Nair, R. P. et al. Sequence and haplotype analysis 161. Czarnowicki, T., Krueger, J. G. & Guttman-Yassky, E.
natural skin oils that function as headless antigens. supports HLA-C as the psoriasis susceptibility 1 gene. Skin barrier and immune dysregulation in atopic
Nat. Immunol. 15, 177–185 (2014). Am. J. Hum. Genet. 78, 827–851 (2006). dermatitis: an evolving story with important clinical
108. de Jong, A. et al. CD1a-autoreactive T cells are a 137. Trembath, R. C. et al. Identification of a major implications. J. Allergy Clin. Immunol. Pract. 2,
normal component of the human alphabeta T cell susceptibility locus on chromosome 6p and evidence 371–379 (2014).
repertoire. Nat. Immunol. 11, 1102–1109 (2010). for further disease loci revealed by a two stage 162. McGrath, J. A. & Uitto, J. The filaggrin story:
109. Bourgeois, E. A. et al. Bee venom processes human genome-wide search in psoriasis. Hum. Mol. Genet. 6, novel insights into skin-barrier function and disease.
skin lipids for presentation by CD1a. J. Exp. Med. 813–820 (1997). Trends Mol. Med. 14, 20–27 (2008).
212, 149–163 (2015). 138. Gottlieb, S. L. et al. Response of psoriasis to a 163. Esaki, H. et al. Early-onset pediatric atopic dermatitis
110. Cheung, K. L. et al. Psoriatic T cells recognize neolipid lymphocyte-selective toxin (DAB389IL-2) suggests a is TH2 but also TH17 polarized in skin. J. Allergy Clin.
antigens generated by mast cell phospholipase primary immune, but not keratinocyte, pathogenic Immunol. 138, 1639–1651 (2016).
delivered by exosomes and presented by CD1a. basis. Nat. Med. 1, 442–447 (1995). 164. Jarrett, R. et al. Filaggrin inhibits generation of
J. Exp. Med. 213, 2399–2412 (2016). 139. Zheng, Y. et al. Interleukin-22, a T(H)17 cytokine, CD1a neolipid antigens by house dust mite-derived
111. Kim, J. H. et al. CD1a on Langerhans cells controls mediates IL-23-induced dermal inflammation and phospholipase. Sci. Transl Med. 8, 325ra318 (2016).
inflammatory skin disease. Nat. Immunol. 17, acanthosis. Nature 445, 648–651 (2007). 165. Gutowska-Owsiak, D., Schaupp, A. L., Salimi, M.,
1159–1166 (2016). 140. Griffiths, C. E. et al. Comparison of ustekinumab and Taylor, S. & Ogg, G. S. Interleukin-22 downregulates
112. Vincent, M. S. et al. CD1-dependent dendritic cell etanercept for moderate-to-severe psoriasis. N. Engl. filaggrin expression and affects expression of
instruction. Nat. Immunol. 3, 1163–1168 (2002). J. Med. 362, 118–128 (2010). profilaggrin processing enzymes. Br. J. Dermatol. 165,
113. Grice, E. A. et al. A diversity profile of the human 141. Lowes, M. A. et al. Psoriasis vulgaris lesions contain 492–498 (2011).
skin microbiota. Genome Res. 18, 1043–1050 discrete populations of Th1 and Th17 T cells. J. Invest. 166. Islam, S. A. et al. Mouse CCL8, a CCR8 agonist,
(2008). Dermatol. 128, 1207–1211 (2008). promotes atopic dermatitis by recruiting IL-5+ T(H)2
114. Grice, E. A. & Segre, J. A. The skin microbiome. 142. Papp, K. A. et al. Brodalumab, an anti-interleukin-17- cells. Nat. Immunol. 12, 167–177 (2011).
Nat. Rev. Microbiol. 9, 244–253 (2011). receptor antibody for psoriasis. N. Engl. J. Med. 366, 167. Soumelis, V. et al. Human epithelial cells trigger
115. Scharschmidt, T. C. et al. Commensal microbes and 1181–1189 (2012). dendritic cell mediated allergic inflammation by
hair follicle morphogenesis coordinately drive Treg 143. Papp, K. et al. Tildrakizumab (MK-3222), an producing TSLP. Nat. Immunol. 3, 673–680 (2002).
migration into neonatal skin. Cell Host Microbe 21, anti-interleukin-23p19 monoclonal antibody, improves 168. Cevikbas, F. et al. A sensory neuron-expressed IL-31
467–477 (2017). psoriasis in a phase IIb randomized placebo-controlled receptor mediates T helper cell-dependent itch:
116. Naik, S. et al. Compartmentalized control of skin trial. Br. J. Dermatol. 173, 930–939 (2015). Involvement of TRPV1 and TRPA1. J. Allergy Clin.
immunity by resident commensals. Science 337, 144. Papp, K. A. et al. Anti-IL-17 receptor antibody AMG Immunol. 133, 448–460 (2014).
1115–1119 (2012). 827 leads to rapid clinical response in subjects with 169. Simpson, E. L. et al. Two phase 3 trials of dupilumab
117. Naik, S. et al. Commensal-dendritic-cell interaction moderate to severe psoriasis: results from a phase I, versus placebo in atopic dermatitis. N. Engl. J. Med.
specifies a unique protective skin immune signature. randomized, placebo-controlled trial. J. Invest. 375, 2335–2348 (2016).
Nature 520, 104–108 (2015). Dermatol. 132, 2466–2469 (2012). 170. Schmitz, J. et al. IL-33, an interleukin-1-like cytokine
118. Saoudi, A., Seddon, B., Heath, V., Fowell, D. & 145. Leonardi, C. et al. Anti-interleukin-17 monoclonal that signals via the IL-1 receptor-related protein ST2
Mason, D. The physiological role of regulatory T cells antibody ixekizumab in chronic plaque psoriasis. and induces T helper type 2-associated cytokines.
in the prevention of autoimmunity: the function of the N. Engl. J. Med. 366, 1190–1199 (2012). Immunity 23, 479–490 (2005).
thymus in the generation of the regulatory T cell 146. Sofen, H. et al. Guselkumab (an IL-23-specific mAb) 171. Lindemans, C. A. et al. Interleukin-22 promotes
subset. Immunol. Rev. 149, 195–216 (1996). demonstrates clinical and molecular response in intestinal-stem-cell-mediated epithelial regeneration.
119. Singh, B. et al. Control of intestinal inflammation by patients with moderate-to-severe psoriasis. J. Allergy Nature 528, 560–564 (2015).
regulatory T cells. Immunol. Rev. 182, 190–200 Clin. Immunol. 133, 1032–1040 (2014). 172. Guttman-Yassky, E. et al. Efficacy and safety of
(2001). 147. Duerr, R. H. et al. A genome-wide association study fezakinumab (an IL-22 monoclonal antibody) in adults
120. Hori, S., Nomura, T. & Sakaguchi, S. Control of identifies IL23R as an inflammatory bowel disease with moderate-to-severe atopic dermatitis
regulatory T cell development by the transcription gene. Science 314, 1461–1463 (2006). inadequately controlled by conventional treatments:
factor Foxp3. Science 299, 1057–1061 (2003). 148. Cargill, M. et al. A large-scale genetic association a randomized, double-blind, phase 2a trial. J. Am.
121. Fontenot, J. D., Gavin, M. A. & Rudensky, A. Y. Foxp3 study confirms IL12B and leads to the identification of Acad. Dermatol. 78, 872–881 (2018).
programs the development and function of CD4+ IL23R as psoriasis-risk genes. Am. J. Hum. Genet. 80, 173. Mirzoyev, S. A., Schrum, A. G., Davis, M. D. P.
CD25+ regulatory T cells. Nat. Immunol. 4, 330–336 273–290 (2007). & Torgerson, R. R. Lifetime incidence risk of alopecia
(2003). 149. Nair, R. P. et al. Genome-wide scan reveals association areata estimated at 2.1% by Rochester Epidemiology
122. Khattri, R., Cox, T., Yasayko, S. A. & Ramsdell, F. of psoriasis with IL-23 and NF-kappaB pathways. Project, 1990–2009. J. Invest. Dermatol. 134,
An essential role for Scurfin in CD4+ CD25+ T Nat. Genet. 41, 199–204 (2009). 1141–1142 (2014).
regulatory cells. Nat. Immunol. 4, 337–342 (2003). 150. Nair, R. P. et al. Polymorphisms of the IL12B and 174. Strazzulla, L. C. et al. Alopecia areata: disease
123. Seneschal, J., Clark, R. A., Gehad, A., IL23R genes are associated with psoriasis. J. Invest. characteristics, clinical evaluation, and new
Baecher-Allan, C. M. & Kupper, T. S. Human epidermal Dermatol. 128, 1653–1661 (2008). perspectives on pathogenesis. J. Am. Acad. Dermatol.
Langerhans cells maintain immune homeostasis in 151. Zaba, L. C., Krueger, J. G. & Lowes, M. A. Resident 78, 1–12 (2018).
skin by activating skin resident regulatory T cells. and “inflammatory” dendritic cells in human skin. 175. Pratt, C. H. et al. Alopecia areata. Nat. Rev. Dis.
Immunity 36, 873–884 (2012). J. Invest. Dermatol. 129, 302–308 (2009). Primers 3, 17011 (2017).
124. Hirahara, K. et al. The majority of human peripheral 152. Wu, C. et al. Induction of pathogenic TH17 cells by 176. Petukhova, L. et al. Genome-wide association study in
blood CD4+CD25highFoxp3+ regulatory T cells bear inducible salt-sensing kinase SGK1. Nature 496, alopecia areata implicates both innate and adaptive
functional skin-homing receptors. J. Immunol. 177, 513–517 (2013). immunity. Nature 466, 113–117 (2010).
4488–4494 (2006). 153. Ciofani, M. et al. A validated regulatory network 177. Betz, R. C. et al. Genome-wide meta-analysis in
125. Rosenblum, M. D. et al. Response to self antigen for Th17 cell specification. Cell 151, 289–303 alopecia areata resolves HLA associations and reveals
imprints regulatory memory in tissues. Nature 480, (2012). two new susceptibility loci. Nat. Commun. 6, 5966
538–542 (2011). 154. Hueber, W. et al. Secukinumab, a human anti-IL-17A (2015).
126. van der Veeken, J. et al. Memory of inflammation in monoclonal antibody, for moderate to severe Crohn’s 178. Gilhar, A., Etzioni, A. & Paus, R. Alopecia areata.
regulatory T cells. Cell 166, 977–990 (2016). disease: unexpected results of a randomised, N. Engl. J. Med. 366, 1515–1525 (2012).

www.nature.com/nri
Reviews

179. Dressel, D. et al. Alopecia areata but not androgenetic 189. Rashighi, M. et al. CXCL10 is critical for the patients with moderate to severe psoriasis: results
alopecia is characterized by a restricted and progression and maintenance of depigmentation in a from the phase III, double-blinded, placebo- and active
oligoclonal T cell receptor-repertoire among infiltrating mouse model of vitiligo. Sci. Transl Med. 6, 223ra223 comparator-controlled VOYAGE 1 trial. J. Am. Acad.
lymphocytes. J. Cutan. Pathol. 24, 164–168 (1997). (2014). Dermatol. 76, 405–417 (2017).
180. Gilhar, A., Ullmann, Y., Berkutzki, T., Assy, B. & 190. Harris, J. E. et al. A mouse model of vitiligo with 199. Papp, K. A. et al. Risankizumab versus ustekinumab
Kalish, R. S. Autoimmune hair loss (alopecia areata) focused epidermal depigmentation requires for moderate-to-severe plaque psoriasis. N. Engl. J.
transferred by T lymphocytes to human scalp explants IFN-gamma for autoreactive CD8+ T cell accumulation Med. 376, 1551–1560 (2017).
on SCID mice. J. Clin. Invest. 101, 62–67 (1998). in the skin. J. Invest. Dermatol. 132, 1869–1876 200. Timerman, D. et al. Novel application of high-dose
181. Xing, L. et al. Alopecia areata is driven by cytotoxic T (2012). rate brachytherapy for severe, recalcitrant
lymphocytes and is reversed by JAK inhibition. 191. Richmond, J. M. et al. CXCR3 depleting antibodies palmoplantar pustulosis. Clin. Exp. Dermatol. 41,
Nat. Med. 20, 1043–1049 (2014). prevent and reverse vitiligo in mice. J. Invest. 498–501 (2016).
182. Liu, L. Y., Craiglow, B. G., Dai, F. & King, B. A. Dermatol. 137, 982–985 (2017).
Tofacitinib for the treatment of severe alopecia areata 192. Harris, J. E. et al. Rapid skin repigmentation on oral Acknowledgements
and variants: a study of 90 patients. J. Am. Acad. ruxolitinib in a patient with coexistent vitiligo and A.W.H. is supported by a Career Development Award from
Dermatol. 76, 22–28 (2017). alopecia areata (AA). J. Am. Acad. Dermatol. 74, the Dermatology Foundation. T.S.K. is supported by grants
183. Strazzulla, L. C. et al. Alopecia areata: an appraisal of 370–371 (2016). R01 AR065807, R01 AI127654 and R01 CA210372 from
new treatment approaches and overview of current 193. Craiglow, B. G. & King, B. A. Tofacitinib citrate for the the US National Institutes of Health.
therapies. J. Am. Acad. Dermatol. 78, 15–24 (2018). treatment of vitiligo: a pathogenesis-directed therapy.
184. Taieb, A. & Picardo, M. Clinical practice. Vitiligo. JAMA Dermatol. 151, 1110–1112 (2015).
Author contributions
N. Engl. J. Med. 360, 160–169 (2009). 194. Rothstein, B. et al. Treatment of vitiligo with the
Both authors contributed to researching data, discussion of
185. Frisoli, M. L. & Harris, J. E. Vitiligo: mechanistic topical Janus kinase inhibitor ruxolitinib. J. Am. Acad.
content and to the writing, review and editing of this article.
insights lead to novel treatments. J. Allergy Clin. Dermatol. 76, 1054–1060 (2017).
Immunol. 140, 654–662 (2017). 195. Cheuk, S. et al. CD49a expression defines tissue-
186. van den Boorn, J. G. et al. Autoimmune destruction of resident CD8+ T cells poised for cytotoxic function in Competing interests
skin melanocytes by perilesional T cells from vitiligo human skin. Immunity 46, 287–300 (2017). The authors declare no competing interests.
patients. J. Invest. Dermatol. 129, 2220–2232 (2009). 196. Malik, B. T. et al. Resident memory T cells in the
187. Jimbow, K., Chen, H., Park, J. S. & Thomas, P. D. skin mediate durable immunity to melanoma. Sci. Publisher’s note
Increased sensitivity of melanocytes to oxidative stress Immunol. 2, eaam6346 (2017). Springer Nature remains neutral with regard to jurisdictional
and abnormal expression of tyrosinase-related protein 197. Schwartz, D. M. et al. JAK inhibition as a therapeutic claims in published maps and institutional affiliations.
in vitiligo. Br. J. Dermatol. 144, 55–65 (2001). strategy for immune and inflammatory diseases.
188. Speeckaert, R. et al. Critical appraisal of the oxidative Nat. Rev. Drug Discov. 17, 78 (2017). Reviewer information
stress pathway in vitiligo: a systematic review and 198. Blauvelt, A. et al. Efficacy and safety of guselkumab, Nature Reviews Immunology thanks P. Scott and the other
meta-analysis. J. Eur. Acad. Dermatol. Venereol. 32, an anti-interleukin-23 monoclonal antibody, compared anonymous reviewer(s) for their contribution to the peer
1089–1098 (2018). with adalimumab for the continuous treatment of review of this work.

Nature Reviews | Immunology

You might also like