You are on page 1of 12

Immunity

Review

Human T Cell Development, Localization,


and Function throughout Life
Brahma V. Kumar,1 Thomas J. Connors,1,4 and Donna L. Farber1,2,3,*
1Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032
2Department of Surgery, Columbia University Medical Center, New York, NY 10032
3Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032
4Department of Pediatrics, Columbia University Medical Center, New York, NY 10032

*Correspondence: df2396@cumc.columbia.edu
https://doi.org/10.1016/j.immuni.2018.01.007

Throughout life, T cells coordinate multiple aspects of adaptive immunity, including responses to pathogens,
allergens, and tumors. In mouse models, the role of T cells is studied in the context of a specific type of path-
ogen, antigen, or disease condition over a limited time frame, whereas in humans, T cells control multiple in-
sults simultaneously throughout the body and maintain immune homeostasis over decades. In this review,
we discuss how human T cells develop and provide essential immune protection at different life stages
and highlight tissue localization and subset delineation as key determinants of the T cell functional role in im-
mune responses. We also discuss how anatomic compartments undergo distinct age-associated changes in
T cell subset composition and function over a lifetime. It is important to consider age and tissue influences on
human T cells when developing targeted strategies to modulate T cell-mediated immunity in vaccines and
immunotherapies.

Introduction proliferation, and differentiation to effector cells that then migrate


The establishment and maintenance of immune responses, ho- to diverse sites to promote pathogen clearance through produc-
meostasis, and memory depends on T cells. T cells express a re- tion of effector cytokines and cytotoxic mediators. Activated
ceptor with the potential to recognize diverse antigens from effector cells are short-lived, although a proportion survive as
pathogens, tumors, and the environment and also maintain memory T cells that persist as heterogeneous subsets based
immunological memory and self-tolerance. T cells are also impli- on migration, tissue localization, and self-renewal capacities.
cated as major drivers of many inflammatory and autoimmune Memory subsets can participate in maintaining long-term immu-
diseases. The in vivo functional role of T cells in immunity and nity and recall protective responses, although their origin and
immunopathology and the underlying mechanisms involved lineage relationship remains unresolved.
have been largely elucidated from mouse models and have led Because humans experience a relatively long lifespan, the crit-
to the development and advancement of immune-based cures ical role of T cells in immunity needs to be studied in the context of
and immunotherapies in humans (Cohen, 2014; Rosenberg, different life stages (Figure 1). In early life (infancy and early child-
2014). However, the power and utility of mouse models to test hood), the majority of T cells are naive T cells newly emerged from
hypotheses depend on reducing the scope of inquiry to one the thymus, although Treg cells are also significantly represented.
type of infection or disease perturbation over a defined time During this formative stage, when the greatest number of new
period under sterile, pathogen-free conditions. By contrast, hu- antigens are encountered, T cells play key protective roles in fend-
mans are continuously exposed to multiple benign and patho- ing off pathogens, Treg cells are critical for developing tolerance to
genic microorganisms and harbor chronic pathogens, yet they innocuous and ubiquitous antigens, and long-term reserves of
can survive for many decades free of major infections even in memory T cells are established. Memory T cells are generated
advanced years (Evans et al., 2014). In order to elucidate mech- from exposure to antigen and begin to accumulate during child-
anisms for the unique longevity and stability of human immunity, hood; the level of memory T cell accumulation then plateaus in
it is important to study T cells within the complex environment of adulthood and is maintained over decades (Saule et al., 2006).
the human body—in multiple sites, at all ages, and across many The change in T cell predominance from naive to memory after
individuals. childhood and the relative stability of immunity over decades of
T lymphocytes originate from bone marrow (BM) progenitors adulthood suggest different roles for T cells in adults than in chil-
that migrate to the thymus for maturation, selection, and subse- dren (Figure 1). In adulthood, fewer new antigens are encoun-
quent export to the periphery. Peripheral T cells comprise tered, and tolerance establishment can be less prevalent, so the
different subsets, including naive T cells, which have the capac- role of T cells shifts to maintaining homeostasis and immunoreg-
ity to respond to new antigens, memory T cells, which derive ulation in the context of repeat and chronically encountered anti-
from previous antigen activation and maintain long-term immu- gens. Surveillance for tumors is also important during this period.
nity, and regulatory T (Treg) cells, which keep immune responses At the later stages of life, well-documented immunosenescent
in check. Immune responses commence when naive T cells changes (for a review, see Goronzy and Weyand, 2017), including
encounter antigen and costimulatory ligands presented by increased inflammation and a decline in T cell functionality,
dendritic cells (DCs), resulting in interleukin 2 (IL-2) production, contribute to immune dysregulation and associated pathology.

202 Immunity 48, February 20, 2018 ª 2018 Elsevier Inc.


Immunity

Review
Figure 1. Overview of the Changing Role of
T Cells at Distinct Life Stages
In early childhood, when humans encounter many
antigens for the first time, T cells mediate path-
ogen clearance for multiple acute infections,
develop memory responses, and establish toler-
ance to innocuous foreign antigens. After child-
hood, the T cell compartment is more stable,
encounters fewer new infections, and generates
less memory. During many decades of adult life,
T cells maintain homeostasis in tissues by con-
trolling chronic infections, surveilling for cancer
cells, and maintaining proper immunoregulation.
Finally, in advanced age there is a well-docu-
mented decline in T cell function and a corre-
sponding increased susceptibility to infection,
cancer, and autoimmunity.

The role of T cells in immune responses and at different life cells, defined as CD4+CD25+ cells expressing the Foxp3 tran-
stages is not uniform across the body. T cells populate virtually scription factor (Hori et al., 2003), represent 9%–10% of human
every organ and tissue, including primary and secondary CD4+ SP thymocytes (Watanabe et al., 2005) and express naive
lymphoid tissue, mucosal and barrier sites, exocrine organs, CD45RA+CCR7+ phenotypes (Seddiki et al., 2006). Most of our
fat, and even the brain and central nervous system. In terms of knowledge of thymopoiesis, the genesis of new T cells and
numbers, the majority of T cells in the human body are found mechanisms for their selection and development, derives from
within lymphoid tissues (BM, spleen, tonsils, and an estimated studies involving thymectomy, BM reconstitution, and genetic
500–700 lymph nodes). Large numbers are also present in manipulation in mice (for reviews, see Germain, 2002 and Klein
mucosal sites (lungs and small and large intestines) and skin et al., 2014). In humans, similar manipulations are not possible;
and an estimated 2%–3% of the total T cell complement is found however, ‘‘natural’’ experiments involving thymectomy and
in human peripheral blood (Clark, 2010; Ganusov and De Boer, thymic transplantation and studies of recent thymic emigrants
2007). In early life, newly generated naive T cells and Treg cells (RTEs) provide key insights into human thymopoiesis and how
populate major lymphoid and mucosal sites in the body, and it differs from thymopoiesis in mice.
memory T cells begin to develop largely in mucosal sites such Unlike mice, who are born lymphopenic with low numbers of
as the small intestine and lung (Thome et al., 2016a). After child- lymphocytes in the periphery, humans are born with a full com-
hood, memory T cells are the predominant subset throughout the plement of T cells (Burt, 2013). In utero, human T cell progenitors
body; however, the accumulation of memory T cells in lymphoid are detected in fetal thymi as early as 9 weeks of gestation, and
tissues occurs at a slower rate during childhood and reaches a mature T cells appear in the thymus by 12–13 weeks and in the
lower maximum frequency than accumulation in mucosal and spleen and lymph nodes (LNs) by 24 weeks of gestation (Haynes
barrier sites (Thome et al., 2014). These findings suggest distinct et al., 1988). Human Treg cells also develop early in fetal life and
roles for T cells in immunity not only at different life stages but are detected in thymi at 12 weeks and in LNs at 14 weeks of
also in specific anatomic compartments. gestation (Cupedo et al., 2005; Michae €lsson et al., 2006). It
In this review, we will consider the development, function, and was established some time ago that neonatal thymectomy in
differentiation of human T cells within the complexities of the mice results in profound immunodeficiency and multi-organ lym-
human condition; such complexities include long-term survival phocytic infiltration (Dalmasso et al., 1963; Sakaguchi et al.,
with multiple environmental, pathogenic, and benign antigens 1982) as a result of defects in the development of naive T cells
through many routes and tissue sites of exposure. We will and Treg cells, respectively (Sakaguchi et al., 1995). In contrast,
discuss these multiple aspects of human T cell responses, neonatal thymectomy in humans, which is performed during car-
beginning with T cell development, the role of naive and regula- diac surgery to repair congenital abnormalities, does not impair
tory T cells, differentiation to effector and memory subsets, and survival or result in deleterious consequences. Adults who
how tissue localization impacts T cell function and maintenance. have undergone neonatal thymectomy (many of whom are now
We will also discuss how all of these aspects of human T cells in their third to fourth decade of life) do not experience increased
function at distinct stages of life, from infancy through adulthood, incidence of infections (Mancebo et al., 2008; Wells et al., 1998),
and how specific sites experience different rates of immunose- and they remain healthy despite having more extensive declines
nescence relating to T cell function and persistence. in naive T cell frequencies with age than control individuals (Pre-
log et al., 2009; van den Broek et al., 2016). Moreover, neonatally
Thymopoiesis thymectomized adults do not have increased incidences of auto-
The thymus, which is the primary site of T cell development, is immunity or allergy when they are compared to aged-matched
where BM progenitors lacking CD4+ and CD8+ coreceptors controls (Silva et al., 2017), consistent with their maintenance
undergo T cell receptor (TCR) rearrangement to generate of normal blood Treg cell frequencies and numbers (Silva et al.,
CD4+CD8+ double-positive (DP) thymocytes. DP cells undergo 2016). However, infants who, as a result of deletion or mutation
selection, giving rise to CD4+ or CD8+ single-positive (SP) thymo- of the Foxp3 transcription factor, are born with a congenital
cytes that ultimately emerge into the periphery as naive T cells defect in Treg cell development, present with immune dysregu-
exhibiting CD45RA+CCR7+ phenotypes. Newly developed Treg lation, polyendocrinopathy, enteropathy, and X-linked (IPEX)

Immunity 48, February 20, 2018 203


Immunity

Review

syndrome manifested by systemic autoimmunity and multi- in individuals over 40 years of age (Douek et al., 1998; Thome
organ infiltrates (Bennett et al., 2001). Therefore, the critical et al., 2016b). Residual thymic activity can persist beyond the
events in T cell development commence prior to birth in humans, fifth decade of life, as shown by the observation that RTE-like
and humans are born with a T cell complement sufficient for anti- cells can be detected in peripheral blood (Jamieson et al.,
pathogen immunity and immunoregulation—at least in the devel- 1999) and in renal transplant recipients after T cell depletion ther-
oped world. apy (Gurkan et al., 2010). Together, these findings suggest that
T cell development in the thymus involves rigorous selection although thymic function effectively ceases after the fourth
events, as determined in mouse models. In mice, positive selec- decade of life, persistence of residual thymus tissue in some in-
tion for self-MHC (major histocompatibility complex) recognition dividuals might be amenable to regeneration therapies to rejuve-
in newly generated DP thymocytes is based on interactions with nate the human immune system.
thymic epithelial cells (non-hematopoietic), and negative selec-
tion removes strongly self-reactive clones through interactions Naive and Treg Cell Maintenance
with thymic DCs. Treg cells undergo a different type of selection In light of declining thymic output, how do adults maintain a func-
and tend to be more self-reactive (Stritesky et al., 2012). It is un- tional and diverse naive T cell repertoire for responding to new
clear whether thymic selection events occur similarly in humans, antigens? Studies examining T cell turnover in human volunteers
but novel insights have emerged from studies of thymus trans- and mice that had been administered deuterated water demon-
plantation—a rare surgery used to reconstitute the T cell strated important species-specific differences regarding thymo-
compartment in infants who have complete DiGeorge syndrome poiesis and naive T cell maintenance. In both young and old
and lack a functional thymus (Hudson et al., 2007). Transplanta- mice, the majority of naive T cells derive from thymic output,
tion of fully allogeneic thymus tissues from unrelated infants re- with minimal peripheral division of naive T cells, whereas in hu-
sulted in thymopoiesis, generation of polyclonal functional naive mans, the majority of naive T cells derive from peripheral turn-
T cells, and anti-pathogen immunity, enabling these individuals over, even in younger adults with active thymic output (den
who otherwise would have died from infections to survive (Mar- Braber et al., 2012; Vrisekoop et al., 2008). These studies also
kert et al., 2010; Markert et al., 2003). Interestingly, thymic recip- showed that although the average lifespan of mouse naive
ients are tolerant of self and the thymic transplant (they require T cells is only 6–10 weeks, in humans, individual naive T cells
no immunosuppression) and also generate Treg cells with can persist for 5–10 years (den Braber et al., 2012; Vrisekoop
diverse repertoires (Chinn et al., 2013; Markert et al., 2010). et al., 2008). The intrinsic long lifespan and turnover of human
Mouse studies show that the generation of functional immunity naive T cells can account for their persistence well into old age.
requires positive selection on thymic epithelial cells (TECs); how- A mechanistic view of human naive T cell maintenance
ever, in human thymic transplants, TECs are of donor origin, yet has emerged from studies that used high-throughput DNA
functional T cells emerge that can respond to antigens presented sequencing for assessing TCR repertoires and from examination
by the host antigen-presenting cells (APCs) (Li et al., 2011). of naive T cells in tissue sites. TCR sequencing can measure
Whether human thymocyte selection is more permissive than se- clonal diversity and quantitate expanded clones. Human naive
lection in mice and occurs through interactions with donor T cells exhibit a highly diverse TCR encompassing up to
epithelium and/or thymic APCs is not known; rules for selection 100 million different specificities (Qi et al., 2014). Naive pheno-
in mice might not apply wholly to humans. type cells in the blood of elderly individuals (70–85 years old) still
How long does thymopoiesis persist in humans? The thymus maintain a diverse repertoire of unique TCRb amino acid se-
is largest at birth, and age-related thymic changes, including a quences that, although reduced in comparision to that of young
reduction in thymic volume, loss of epithelial cells, increase in adults (Qi et al., 2014), suggests that humans are capable of re-
perivascular space, and replacement of thymic tissue by fat, sponding to novel antigens even at advanced ages. Study of
begin during childhood (Haynes et al., 2000). Thymic function naive T cells in circulation and multiple tissue sites obtained
can be assessed in peripheral T cells on the basis of markers from organ donors has enabled a novel assessment of how naive
of new thymic emigrants; such markers include surface CD31 T cells are maintained over a lifespan (Thome et al., 2016a;
expression (Junge et al., 2007; Tanaskovic et al., 2010) and Thome et al., 2016b; Thome et al., 2014). Naive T cells comprise
TCR excision circles (TRECs) as products of gene rearrange- a significant proportion (20%–50%) of total T cells within multiple
ment (Hazenberg et al., 2002; Hazenberg et al., 2001). There lymph nodes for decades after cessation of thymic output. Naive
are well-documented decreases in CD31 expression and T cells also maintain functionality as individuals age; no overt
TREC content in peripheral blood T cells as individuals age functional differences exist between naive T cells from younger
(Douek et al., 1998; Jamieson et al., 1999; Junge et al., 2007). individuals with active thymic output and older individuals with
The greatest decline in thymopoiesis was generally believed to no thymic output (Thome et al., 2016b). TCR repertoire analysis
occur during puberty; however, more recent studies suggest of lymphoid, naive, and memory T cells via CDR3 sequencing re-
that this steep reduction could occur later in life. In human thymic vealed high diversity (and corresponding low clonality) of CD4+
tissue obtained from organ donors aged several months to and CD8+ naive T cells compared to the corresponding memory
>70 years, DP thymocytes (indicating ongoing selection) were subset in the spleen and multiple LNs (Thome et al., 2016b), indi-
detected at the expected frequency (60%–80%) in active cating that naive T cells in tissues maintain their capacity to
thymus tissue from donors <40 years of age, whereas few DP respond to diverse antigens.
cells were detected in thymus tissues of adults >40 years of Treg cells also emerge from the thymus and exist at high fre-
age (Thome et al., 2016b). Similarly, TREC levels of naive quencies in tissues during early life; however, their persistence
T cells from human blood, spleen, and LNs were steeply reduced and frequency differs from those of naive T cells. The early

204 Immunity 48, February 20, 2018


Immunity

Review

establishment of Treg cells in utero is followed by elevated levels 14–21 days (Akondy et al., 2015; Blom et al., 2013; Miller et al.,
in early life, when 10%–30% of all CD4+T cells in blood, lymphoid 2008; Wieten et al., 2016a). As in mouse infection models, in hu-
tissue, and mucosal sites are Treg cells, whereas less than 5% mans the magnitude of the effector CD8+ T cell response corre-
are Treg cells in adults (Mold et al., 2008; Thome et al., 2016a). lates to the initial viral load (Akondy et al., 2015) and contracts
Despite differences in overall frequency, pediatric and adult rapidly; it returns to near baseline levels by 30 days (Akondy
Treg cells share similar features. Treg cells maintain a TCR reper- et al., 2015, Kohler et al., 2012, Miller et al., 2008). Virus-specific
toire entirely distinct from corresponding non-Treg cell popula- memory T cells subsequently persist in frequencies reduced
tions, and there is minimal overlap of TCRb sequences between from the effector response (5%–6%) (DeWitt et al., 2015; Miller
these subsets in umbilical-cord blood and in adult peripheral et al., 2008; Wieten et al., 2016b) and are detectable 25 years
blood (Golding et al., 2017). In both adults and children, Treg after vaccination (Fuertes Marraco et al., 2015; Wieten et al.,
cells in blood and lymphoid tissues are CD45RA+CCR7+, indi- 2016b). Similar kinetics of effector expansion and memory
cating a naive phenotype, whereas like conventional memory development were observed after smallpox (vaccinia) and influ-
T cells, Treg cells in mucosal sites are CD45RA and express enza vaccination (Miller et al., 2008); vaccinia-specific memory
CD45RO (Thome et al., 2016a). Functionally, pediatric Treg cells T cells were also detected after many decades (Hammarlund
in the LNs can express higher levels of FoxP3 than adult LN Treg et al., 2003). Together, these studies indicate that human effector
cells (Thome et al., 2016a); however, more studies are needed to T cell expansion, contraction, and memory formation after viral
assess how early-life Treg cells might differ from those that infection are similar in kinetics and magnitude to those of many
persist in later life. acute viruses investigated in mice.
The decline in Treg cell frequency in humans begins earlier in Although the majority of effector T cells contract rapidly and
childhood than the decline in conventional naive populations are not present in significant proportions at steady state, a pop-
(Thome et al., 2016a), suggesting that both thymic and peripheral ulation of terminal effector cells (Temra cells) exhibiting
mechanisms are at play. Studies of mice along with complemen- CD45RA+CCR7 phenotypes can persist in circulation. Temra
tary analyses in humans indicate that age-associated reductions cells are mostly present within the CD8+ T cell lineage and exhibit
in Treg cell generation occur as a result of interactions between a high capacity for IFNg production and a low proliferative ca-
thymic output, peripheral induction, and maintenance. In mice, pacity, and their frequency in blood is correlated with persistent
mature Treg cells migrate back to the thymus and suppress cytomegalovirus (CMV) infection (Larbi and Fulop, 2014). The
thymic production of Treg cells but not conventional naive proportion of CD8+ Temra cells in blood and BM increases
T cells (Thiault et al., 2015). Mature Treg cells were also detected with age, specifically in CMV-seropositive donors (Di Benedetto
in human pediatric thymi (Thiault et al., 2015), suggesting that a et al., 2015; Gordon et al., 2017), suggesting a role for Temra
similar mechanism controls Treg cell production in humans. cells in persistent infections. CD4+ Temra cells are rarely de-
Compared with naive T cells, human Treg cells exhibit higher tected, but expansion of CD4+ Temra cells with cytotoxic func-
turnover, as measured by Ki67 expression (Silva et al., 2016; tion occurs in individuals infected with Dengue virus and is
Thome et al., 2016a), which could also contribute to their associated with protection (Weiskopf et al., 2015). These find-
declining frequency with age. Treg cell frequency might also be ings suggest that certain viruses trigger terminal effector differ-
affected by changes in DC populations. In a recent study, human entiation and Temra cell formation, which might relate to antigen
fetal DCs were shown to promote Treg cell induction more load or persistence. Interestingly, Temra cells appear to be spe-
readily than adult DCs (McGovern et al., 2017). Together, these cific to humans, with no clear correlate in mouse infection
results suggest multiple interacting mechanisms for controlling models.
Treg cell frequencies after childhood and are consistent with
the view that the optimal window for tolerance induction occurs Memory T Cell Heterogeneity and Tissue Distribution
early in life. Memory T cells represent a major circulating population in
human blood and are subdivided on the basis of phenotype
T Cell Effector and Memory Differentiation into central-memory (Tcm, CD45RA CCR7+), effector-memory
Peripheral T cell differentiation has been extensively character- (Tem, CD45RA CCR7 ), and stem-cell memory (Tscm,
ized in mouse infection models and occurs in three phases: CD45RA+CCR7+CD95+CD122+) T cells (Gattinoni et al., 2011;
clonal expansion, in which activated pathogen-specific T cells Sallusto et al., 1999). Functionally, both Tcm and Tem cells are
expand and differentiate into effector T cells that mediate infec- capable of producing IL-2 and effector cytokines upon stimula-
tion clearance; contraction, in which the majority of effector tion; however, Tcm cells exhibit lymphoid homing profiles and
T cells die by apoptosis after an infection; and a memory phase, a high proliferative capacity, whereas Tem cells produce more
in which a fraction of these primed T cells persist as long-term effector cytokines (Gattinoni et al., 2011; Sallusto et al., 1999;
memory T cells that protect against subsequent infection (Gour- Willinger et al., 2005). Tscm cells are a relatively rare subset
ley et al., 2004; Kaech and Wherry, 2007). Studying T cell differ- with high proliferative and self-renewal capabilities, but no
entiation to effector and memory T cell fates in humans requires effector function (Gattinoni et al., 2011). Models for the linear
specific cohorts that are challenged and followed over time. and bifurcated generation of human memory T cell subsets
Elegant studies that used the live-attenuated Yellow fever virus have been previously inferred from functional analysis, differen-
vaccination (YFV-7D) show activated CD4+ and CD8+ T cells tiation markers, and analysis of cell division (Ahmed et al., 2009).
co-expressing CD38 and HLA-DR, which are detectable in the However, recent studies that used epigenetic and transcriptional
blood within 2–3 days; YFV-specific CD8+ T cells are subse- analysis have provided new insights into their differentiation
quently detectable in blood and exhibit peak responses after hierarchy. For CD4+T cells, epigenomic profiling based on

Immunity 48, February 20, 2018 205


Immunity

Review

assessment of methylation status, DNA accessibility, and his- by the fact that mouse Trm cells can be generated in response
tone modifications revealed a progressive loss of DNA methyl- to vaccines, non-infectious allergens, autoantigens, and tumors
ation of key genes that control memory development, in the (Hondowicz et al., 2016; Morawski et al., 2017; Shin and Iwasaki,
order of genes encoding naive—Tcm—Tem cells (Durek et al., 2012; Zens et al., 2016).
2016). A linear progression of differentiation for human CD8+ Phenotypically, mouse Trm cells express the early T cell acti-
T cells in the order naive—Tscm—Tcm—Tem cells was sug- vation marker CD69 (Anderson et al., 2014; Mueller and Mackay,
gested by methylation analysis and chromatin accessibility (Ab- 2016; Turner et al., 2014), which can promote retention through
delsamed et al., 2017; Moskowitz et al., 2017). However, these sequestration of the sphingosine-1-phosphate receptor (S1PR1)
lineage analyses are based on the fraction and subsets of (Mackay et al., 2015; Shiow et al., 2006). CD8+ Trm cells in skin
T cells that emerge into circulation and not on the majority pop- and mucosal sites co-express CD103 in addition to CD69
ulations contained within lymphoid and peripheral tissue sites. (Bergsbaken and Bevan, 2015; Mueller and Mackay, 2016).
The identification of heterogeneous tissue distribution and Although CD69 expression is a common feature of both CD4+
distinct features of T cells in specific sites in mouse models (Bin- and CD8+ Trm cells, CD69 Trm cells defined on the basis of tis-
gaman et al., 2005; Masopust et al., 2001; Masopust et al., 2004) sue retention properties have been identified in mouse tissues
suggested that in humans, circulating T cells might not be repre- (Steinert et al., 2015). Whether CD69 is required for Trm cell for-
sentative of their tissue counterparts. However, human tissue mation also remains a matter of debate; CD69-deficient CD8+
samples available for research purposes are often limited to sur- T cells exhibit reduced Trm cell formation in skin (Mackay
gical explants from disease-affected patients or isolated bi- et al., 2015), whereas CD69 / CD4+ T cells can generate Trm
opsies, and they are derived from low numbers of individuals. cells in lymphoid sites (Ugur et al., 2014).
More recently, the use of organ donor tissues has enabled Recent studies have identified cells with a Trm phenotype in
assessment of T cell subset distribution, compartmentalization, multiple human tissues. Analysis of memory T cells from healthy
and function across blood and multiple lymphoid and mucosal organ donors and tissue explants revealed that CD69 is ex-
sites in healthy humans of all ages (Carpenter et al., 2018; Satha- pressed by the majority of CD4+ and CD8+ memory T cells in
liyawala et al., 2013; Thome et al., 2014). Analysis of T cell sub- multiple sites, including mucosal tissues (lungs, small and large
sets in tissues over six decades of adult life revealed that Tem intestines, and genital mucosa), skin, lymphoid sites (spleen,
cells were the predominant population in the lungs, small and LN, and BM), and exocrine tissues (pancreas and salivary
large intestines, spleen, and blood, as well as in 30%–50% of glands), whereas blood memory T cells are largely CD69 (Booth
the LN T cell complement, whereas naive T cells were primarily et al., 2014; Clark et al., 2006; Gordon et al., 2017; Kumar et al.,
found in the blood, spleen, and LNs (Sathaliyawala et al., 2013; 2017; Okhrimenko et al., 2014; Pallett et al., 2017; Radenkovic
Thome et al., 2014). For CD4+ T cells, Tcm cells are consistently et al., 2017; Swaims-Kohlmeier et al., 2016; Thome et al.,
present in lymphoid sites (average frequency 20%) and at lower 2014; Trimble et al., 2010; Watanabe et al., 2015; Wong et al.,
frequencies in mucosal tissues, whereas CD4+ Temra cells are 2016; Woon et al., 2016). CD103 is expressed by a subset of
rarely present in any tissue (Thome et al., 2014). For CD8+ CD69+ memory CD8+ T cells in barrier and oral-gastro mucosal
T cells, significant fractions (30%) of Temra cells were found sites, but it is not expressed significantly by lymphoid memory
within blood and blood-rich tissues (spleen, BM, and lung) but CD8+ T cells or by CD4+ memory T cells in any tissue (Kumar
not in other sites, and only low frequencies of CD8+ Tcm-pheno- et al., 2017; Thome et al., 2014; Watanabe et al., 2015), suggest-
type cells were found in blood and lymphoid sites (Gordon et al., ing that CD69 is the primary marker distinguishing tissue from
2017; Thome et al., 2014). Notably, these subset frequencies circulating memory T cells. Although CD69 can be upregulated
were quite stable across highly diverse donors, suggesting tis- after T cell activation (Cibrián and Sánchez-Madrid, 2017),
sue-specific compartmentalization of human T cell subsets. CD69+ memory T cells in tissues do not exhibit activation fea-
tures such as HLA-DR, CD38, or CD25 expression (Kumar
Tissue-Resident Memory T (Trm) Cells et al., 2017; Okhrimenko et al., 2014; Thome et al., 2014; Woon
It has recently been established in mouse models that a large et al., 2016). Together, these findings indicate that, in addition
fraction of memory T cells in tissues comprise a distinct subset, to expressing CD69 and, in some cases, CD103, human tissue
designated tissue-resident memory T (Trm) cells (for reviews memory T cells exhibit features of resting memory T cells.
about mouse Trm cells, see Mueller and Mackay, 2016 and Defining human Trm as a distinct subset is supported by tran-
Schenkel and Masopust, 2014). Mouse Trm cells have been scriptional profiling studies of phenotypic subsets. Two recent
described in diverse types of tissues, including liver, brain, and studies showed that human tissue memory T cells expressing
mucosal tissues and tissues from barrier sites and salivary the Trm cell markers CD69 and/or CD103 are a transcriptionally
glands, where in all cases they are retained and do not recircu- distinct human memory subset, with key homologies to mouse
late, as shown by parabiosis and in vivo labeling studies (Jiang Trm cells (Hombrink et al., 2016; Kumar et al., 2017). Notably,
et al., 2012; Mueller and Mackay, 2016; Steinert et al., 2015; Tei- a core signature defining human Trm cells has been established
jaro et al., 2011). Functionally, mouse Trm cells mediate rapid through whole-transcriptome profiling combined with pheno-
in situ protection against diverse viral, bacterial, and parasite in- typic and functional validations in multiple tissue sites for both
fections and are more effective than circulating memory T cells in CD4+ and CD8+ T cells (Kumar et al., 2017) (Figure 2). Human
pathogen clearance (Fernandez-Ruiz et al., 2016; Gebhardt CD69+ (or CD103+) tissue memory T cells and mouse Trm cells
et al., 2009; Glennie et al., 2015; Jiang et al., 2012; Sakai et al., share key transcriptional features, including downregulation of
2014; Schenkel et al., 2014; Teijaro et al., 2011; Thom et al., the homing receptor S1PR1 and its associated transcription fac-
2015). The clinical importance of Trm cells is also underscored tor KLF2 (Hombrink et al., 2016; Kumar et al., 2017; Woon et al.,

206 Immunity 48, February 20, 2018


Immunity

Review
Figure 2. Defining Features of Human Trm
Cells
Homing Regulation Trm cells have a unique profile, which includes
expression of specific transcription factors, hom-
CXCR6 PD-1, LAG3, CTLA4 ing receptors, and adhesion markers to maintain
S1PR1, S1PR5, IL-10 tissue residency and the functional capacity to
CD62L, CX3CR1 Ki67 secrete both pro- and anti-inflammatory cyto-

TRM
kines. Trm cells also upregulate a number of
inhibitory genes and exhibit reduced proliferative
turnover compared to turnover in circulating
memory T cell counterparts.

Notch --> RBPJ


Eomes, T-bet,
KLF2, KLF3
Trm cells from human skin found that the
Retention Function CD49a+CD103+ subset of CD8+ Trm cells
CD103 IL-2 represented a subset with superior cyto-
CD49a IFN-γ toxic abilities compared with those of
CD69 TNF-α their CD49a– counterparts (Cheuk et al.,
IL-17
2017). Furthermore, Trm cells might
adopt tissue-specific functional or migra-
tion capacities, which will be an important
area for future studies.
2016), shown to be required for Trm cell formation in mice (Skon Human Trm cells most likely have key roles in mediating
et al., 2013). Like mouse Trm cells, human Trm cells exhibit up- protective responses and maintaining long-term immunity. Indi-
regulation of CXCR6 and ITGA1 (CD49a), a collagen-binding in- rect evidence that Trm cells are the critical protective subset
tegrin, and downregulation of SELL (CD62L) and CX3CR1 and derives from studies of patients given alemtuzumab, an agent
thus avoid egress cues (Hombrink et al., 2016; Kumar et al., that profoundly depletes circulating T cells while preserving
2017; Wong et al., 2016). However, the transcriptional regulators Trm cells as assessed in skin (Clark et al., 2012). Remarkably,
of human and mouse Trm cells might be different. In mice, the these patients do not experience high rates of infection (Clark
transcription factor Hobit is exclusively expressed by and et al., 2012), and transplant recipients, who are also treated
required for the development of Trm cells after infection (Mackay with depletional agents and other types of systemic immuno-
et al., 2016), whereas in humans, Trm phenotype cells express suppression, do not succumb to infections, suggesting that
low to negligible levels of Hobit (ZNF683) transcripts (Kumar memory T cells residing within tissues can adequately control
et al., 2017; Vieira Braga et al., 2015). ongoing and new infections. Furthermore, the presence of
Functional aspects of the core signature reveal a dual role for CD8+ resident T cells in human skin is associated with
human Trm cells; this role encompasses both protection and immune responses to herpes simplex virus (HSV) (Zhu et al.,
regulation (Figure 2). Human Trm cells produce higher levels of 2007; Zhu et al., 2013), analogous to findings in mice that
IFN-g, IL-17, TNF-a, IL-2, and other cytokines associated with showed skin Trm-mediated protection to HSV (Gebhardt
protective immunity than do circulating Tem cells, but they et al., 2009; Gebhardt et al., 2011). These early findings sug-
also produce higher levels of the immunoregulatory cytokine gest that targeting Trm cells could be an effective strategy to
IL-10 (Hombrink et al., 2016; Kumar et al., 2017; Pallett et al., promote vaccine-mediated protection, as supported by prom-
2017; Watanabe et al., 2015). Moreover, human Trm cells ex- ising results in mouse models (Shin and Iwasaki, 2012; Zens
press surface receptors known to potently inhibit T cell function; et al., 2016).
such receptors include PD-1, LAG3, and CTLA-4 (Hombrink One potential mechanism for how human Trm cells might be
et al., 2016; Kumar et al., 2017; Woon et al., 2016), and also optimized for protection is in their maintenance of T cell clones
CD101 (Kumar et al., 2017), a molecule associated with inhibition specific to pathogens encountered at their site of residence.
of proliferation (Soares et al., 1998). Interestingly, human Trm For example, studies have found biased maintenance of influ-
cells also exhibit lower expression of the proliferation marker enza-specific CD8+ T cells within the human lung Trm subset
Ki67 than do circulating Tem cells (Kumar et al., 2017). We pro- (Purwar et al., 2011; Turner et al., 2014), hepatitis B virus-specific
pose that these dual protective and regulatory capabilities are CD8+ T cells within liver CD69+ memory T cells (Pallett et al.,
critical for long-term maintenance; Trm cells exist in a quiescent 2017), and EBV-specific CD8+ Trm cells in the spleen and tonsils
state to promote longevity and have anti-inflammatory and reg- (Woon et al., 2016). The tissue distribution of T cells specific for
ulatory function to prevent unnecessary activation and tissue systemic viruses that infect and/or persist in multiple sites is
damage, but they also retain the ability to respond quickly more complex. CMV-specific T cells exhibit different distribution
upon pathogen invasion. patterns between individuals but are predominantly found in
In addition to the core signature, there is evidence for hetero- blood, BM (majority of donors), lung, and LNs. (Gordon et al.,
geneity within human Trm cells. CD103 expression on human 2017). BM was also found to be enriched in comparison to blood
CD8+ Trm cells can vary, and in mice, CD103+ and CD103 for specificities to multiple systemic pathogens (Okhrimenko
Trm cells are developmentally distinct subsets (Bergsbaken et al., 2014), suggesting compartmentalization of long-lived
and Bevan, 2015; Kumar et al., 2017). A recent study examining memory populations in the BM. Taken together, these findings

Immunity 48, February 20, 2018 207


Immunity

Review

suggest specific generation and/or retention of memory T cells at T Cell Subsets over the Human Lifespan
the infection site. As described in the sections above, human T cell development,
In humans, a lifetime of exposure to pathogens and differentiation, and maintenance are all dynamic processes that
commensal microbiota most likely has a profound impact on change over the course of human life. Combining T cell subset
the development and maintenance of Trm cells in ways that do frequency with age along with anatomic localization of the cells,
not occur in conventional mouse models. For example, the frac- as diagrammed schematically in Figure 3, reveals that tissue
tion of Trm-phenotype cells in tissues is consistently higher in hu- sites exhibit different qualitative and quantitative changes in
mans than in mice, and memory T cells with a canonical Trm cell the T cell compartment with age. The thymus is the immune or-
phenotype (CD69+CD49a+CD103+) can be found in human gan exhibiting the earliest functional role in production of new
lymphoid tissue (Kumar et al., 2017) but are not typically present T cells during fetal life, yet its rate of decline is faster than that
in mouse lymphoid sites. The cumulative effects of multiple path- of other lymphoid sites. Notably, the contribution of thymopoie-
ogen exposure on establishment of Trm cells in tissues is also sis to new naive T cells (and Treg cells) is highest during infancy
suggested by studies of ‘‘dirty’’ pet store mice, which have and declines in early childhood as active thymus tissue is con-
higher frequencies of CD69+ T cells within tissues than conven- verted to fat. During adulthood, thymic output is very low and
tional mice in spf (specific pathogen free) facilities (Beura et al., continues until the fourth or fifth decade of life, when it ceases
2016). In addition, the accumulation of Trm cells with age occurs altogether.
more rapidly in intestines, followed by lungs (Thome et al., 2014), In the periphery, new naive T cells emerging from the thymus
where multiple commensal organisms are also present. Taken populate blood and multiple mucosal and lymphoid tissue sites
together, these results suggest that the rate and extent of Trm in early life (Thome et al., 2016a). These sites exhibit both global
cell establishment in tissues could be directly related to antigen and tissue-specific changes in T cell subset composition as indi-
load at specific sites. viduals age (Figure 3). In all sites, there is a progressive reduction
in naive T cells along with a compensatory increase in memory
Tissue Residency of Additional T Cell Subsets subsets from childhood throughout adulthood; however, the
Tissue residency might not be unique to memory T cells. There is rate of this change is fastest in mucosal sites, intermediate in
evidence that naive T cells can persist in tissues, and particularly the spleen, and slowest in the lymph nodes and blood. Notably,
within LNs. Clonal analysis of naive T cells in spleen and LNs of early memory T cells appear primarily in intestines and lungs, and
individuals revealed no overlap between the TCR repertoires of memory T cells predominate in these sites by late childhood
naive T cells from different lymphoid sites, regardless of age (Figure 3). These mucosal memory T cells exhibit Trm cell pheno-
(Thome et al., 2016b). Even naive T cell clones that were types, which develop gradually in the first two years of life but
modestly expanded were largely limited to a single lymphoid rapidly accumulate to become the predominant subset
site, in contrast with expanded memory populations, which throughout adulthood. In the spleen and lymph nodes, the loss
showed high overlap between sites (Thome et al., 2016b). These of naive cells and accumulation of memory cells with age is
data suggest that naive T cells take up long-term residence in more gradual; an average of 40%–50% of memory T cells adopt
lymph nodes, where they can expand in situ, such that lymph Trm cell properties in these sites. However, compared to spleen
nodes serve as reservoirs for their maintenance. Given that naive and blood, the LNs exhibit the slowest decline in naive T cell fre-
T cells lack expression of canonical Trm cell markers, we pro- quency and accumulation of memory T cells with age (Thome
pose that retention mechanisms for naive T cells in LNs might et al., 2016b) (Saule et al., 2006). CD4+ Treg cells in blood and
be more dependent on cytokine signaling rather than specific tissue sites are highest in frequency during early life; they decline
cell-cell interactions. during childhood, and adults maintain relatively low, but stable,
The concept of tissue residency has also been described for proportions (1%–8%) in blood and tissues (Thome et al.,
Treg cells. In mice, tissue Treg cells in fat, lung, and muscle serve 2016a). CD8+ Temra cell populations tend to increase in blood
key roles in metabolic homeostasis and tissue repair and exhibit and blood-rich sites such as BM, spleen, and lungs as individ-
distinct phenotypes and transcriptional profiles compared to uals age, and individuals with persistent CMV infection show a
those of lymphoid Treg cells (for a review, see Panduro et al., steeper increase (Gordon et al., 2017) (Figure 3). Thus, age-asso-
2016). In human tissues, a proportion of Treg cells (40%–50%) ciated changes in the T cell compartment exhibit site-specific
express the Trm cell marker CD69 (Thome et al., 2016a). Treg and subset-specific dynamics.
cells residing in human skin also express skin-homing receptors What are the potential implications of this differential immuno-
CLA, CCR4, and CCR6 (Clark and Kupper, 2007). Higher propor- logical aging of T cells in various sites? The rapid accumulation of
tions of Treg cells that differentially produce IL-17 have been memory T cells in mucosal sites most likely reflects the high an-
identified in psoriatic skin lesions than in unaffected skin (San- tigen load encountered, particularly for new antigens during in-
chez Rodriguez et al., 2014), suggesting roles for Treg cells in fancy and early childhood. The spleen is also a site for diverse
controlling local homeostasis. Treg cells have also been identi- antigen encounter via bloodborne antigens, and it accumulates
fied in human fat, where reduced numbers of Treg cells have both circulating and resident memory T cells. By contrast, lymph
been correlated with obesity (Feuerer et al., 2009). Human tissue nodes, which receive antigenic signals via DCs migrating from
Treg cells in LNs could preserve homeostasis, given that their tissues, are likely to experience lower antigen loads than other
depletion results in increased T cell proliferation and cytokine sites. The early accumulation of Trm cells in mucosal and barrier
production ex vivo (Peters et al., 2013; Thome et al., 2016a). Dis- sites can act to control antigen load and inflammation and in turn
secting the importance and functional role of human tissue Treg limit DC maturation and migration to draining LNs. In this way,
cells in maintaining homeostasis will require further studies. in situ responses in the sites of pathogen entry prevent LN

208 Immunity 48, February 20, 2018


Immunity

Review
Figure 3. Distinct Changes in Human T Cell
Stage: Infant Child Youth Adult Senior Subsets in Diverse Tissue Sites as
Individuals Age
The T cell compartment in humans undergoes
major changes from infancy to old age, and
Thymus Thymic contribution key differences are seen across tissue sites.
to naïve pool Thymic output
Thymic output declines from a very early age,
and by middle adulthood there is negligible
thymic output as measured by peripheral
Memory TREC content or DP thymocytes. After early
Naïve +CMV
Blood TEMRA (CD8+) childhood, the relative contribution of the
thymus to the peripheral pool of naive T cells is
Tregs
low as a result of peripheral mechanisms for
TRM naive T cell maintenance. In all tissues, there is
Lymph Memory a shift from naive to memory predominance with
Naïv
Naïve
ï e
Nodes age. This transition occurs during childhood
Tregs
in barrier tissues such as the lungs and intestines,
and much later in adulthood in lymphoid
TRM sites; lymph nodes exhibit the slowest transition
+CMV to memory T cell predominance. Resident
Spleen Memory
Naïve TEMRA (CD8+) memory T cells comprise a varying fraction of the
memory compartment in tissues (highest in bar-
Tregs
rier tissues, lower in lymph nodes); however, the
development of Trm cells lags slightly behind the
TRM
+CMV
development of memory. Terminally differenti-
Lungs Memory ated T cells that re-express CD45RA (Temra)
Naïve TEMRA (CD8+) comprise an important fraction of CD8+ T cells in
Tregs
Naïve the blood, spleen, and lungs, and increased fre-
quencies are seen in old age and with CMV
TRM Memory infection.
Intestines Memory
TRM
Tregs Tregs
Age: 0-2 2-15 15-30 30-70 70+ TEMRA

involvement and LNs therefore provide a protective tissue niche studies will be important for understanding mechanisms for con-
for maintenance of naive and resting memory populations. Evi- trolling T cell homeostasis at different life stages.
dence from mouse infection models further suggest that Trm
cells can promote a generalized anti-pathogen environment in Concluding Remarks
the tissue by local cytokine production and immune cell activa- Although traditional studies of human T cells have focused on the
tion (Ariotti et al., 2014; Schenkel et al., 2014). It is possible blood as the most highly accessible site, more recent studies
that the high Trm cell content in mucosal and barrier tissues dur- have revealed profound anatomic compartmentalization of
ing adulthood promotes a similar protective environment and T cell subsets. Notably, newly defined subsets of tissue-resident
limits pathogen spread without the need for input from circu- memory T cells and tissue localization of other subsets indicate
lating and lymphoid reservoirs. This local maintenance of tissue anatomic complexity of the T cell response. T cell subsets have
immunity might be the reason that adults can tolerate immuno- specialized functions that are associated with the life stage and
suppression for transplantation and chemotherapy for cancer anatomic compartment, and different sites exhibit distinct ki-
without succumbing to multiple infections. netics of changes as individuals age. Molecular, phenotypic,
Site-specific and temporal dynamics in Treg cells might also and functional profiling on the single-cell level will be an impor-
impact immunoregulation at different life stages. The higher tant focus if future studies are to dissect how alterations in
prevalence of Treg cells in young children compared to in older tissue-based immunity might promote or be associated with dis-
children and young adults suggests a distinct early role for immu- ease processes, including those of autoimmune and inflamma-
noregulation during a period of new antigen exposure. After this tory diseases and cancer. Understanding mechanisms for tissue
initial immunoregulatory phase, memory T cells and, specifically, specialization of T cell responses, including potential epigenetic
Trm cells in tissues might also provide regulatory roles, as Trm changes that are driven by the tissue environment, will also
cells exhibit regulatory capacities such as IL-10 production reveal how long-term health and immune homeostasis are main-
and low proliferation (Kumar et al., 2017). The role of Treg cells tained in the face of diverse antigens and persistent or newly
in adult immune responses is challenging to assess because fre- encountered pathogens that are an integral part of the human
quency estimates in disease rely only on circulating Treg cells; condition.
however, altered frequencies and functionality of circulating
Treg cells have been associated with human autoimmune dis- ACKNOWLEDGMENTS
eases, including type 1 diabetes, multiple sclerosis, rheumatoid
arthritis, and inflammatory bowel disease (for reviews see Bac- This work was supported by NIH grants AI106697 and AI128949 awarded
to D.L.F. We wish to thank Michelle Miron for critical reading of this manu-
chetta et al., 2007 and Dejaco et al., 2006). Analysis of tissue- script and Dr. Tomer Granot, Ms. Elana Bell, and Joseph Bell for help with
specific regulation by Trm cells and tissue Treg cells in future the figures.

Immunity 48, February 20, 2018 209


Immunity

Review
REFERENCES Clark, R.A. (2010). Skin-resident T cells: the ups and downs of on site immu-
nity. J. Invest. Dermatol. 130, 362–370.
Abdelsamed, H.A., Moustaki, A., Fan, Y., Dogra, P., Ghoneim, H.E., Zebley,
Clark, R.A., and Kupper, T.S. (2007). IL-15 and dermal fibroblasts induce pro-
C.C., Triplett, B.M., Sekaly, R.P., and Youngblood, B. (2017). Human memory
liferation of natural regulatory T cells isolated from human skin. Blood 109,
CD8 T cell effector potential is epigenetically preserved during in vivo homeo-
194–202.
stasis. J. Exp. Med. 214, 1593–1606.
Clark, R.A., Chong, B., Mirchandani, N., Brinster, N.K., Yamanaka, K., Dow-
Ahmed, R., Bevan, M.J., Reiner, S.L., and Fearon, D.T. (2009). The precursors
giert, R.K., and Kupper, T.S. (2006). The vast majority of CLA+ T cells are resi-
of memory: models and controversies. Nat. Rev. Immunol. 9, 662–668.
dent in normal skin. J. Immunol. 176, 4431–4439.
Akondy, R.S., Johnson, P.L., Nakaya, H.I., Edupuganti, S., Mulligan, M.J.,
Lawson, B., Miller, J.D., Pulendran, B., Antia, R., and Ahmed, R. (2015). Initial Clark, R.A., Watanabe, R., Teague, J.E., Schlapbach, C., Tawa, M.C., Adams,
viral load determines the magnitude of the human CD8 T cell response to yel- N., Dorosario, A.A., Chaney, K.S., Cutler, C.S., Leboeuf, N.R., et al. (2012).
low fever vaccination. Proc. Natl. Acad. Sci. USA 112, 3050–3055. Skin effector memory T cells do not recirculate and provide immune protection
in alemtuzumab-treated CTCL patients. Sci. Transl. Med. 4, 117ra7.
Anderson, K.G., Mayer-Barber, K., Sung, H., Beura, L., James, B.R., Taylor,
J.J., Qunaj, L., Griffith, T.S., Vezys, V., Barber, D.L., and Masopust, D. Cohen, I.R. (2014). Activation of benign autoimmunity as both tumor and auto-
(2014). Intravascular staining for discrimination of vascular and tissue leuko- immune disease immunotherapy: a comprehensive review. J. Autoimmun. 54,
cytes. Nat. Protoc. 9, 209–222. 112–117.

Ariotti, S., Hogenbirk, M.A., Dijkgraaf, F.E., Visser, L.L., Hoekstra, M.E., Song, Cupedo, T., Nagasawa, M., Weijer, K., Blom, B., and Spits, H. (2005). Develop-
J.Y., Jacobs, H., Haanen, J.B., and Schumacher, T.N. (2014). T cell memory. ment and activation of regulatory T cells in the human fetus. Eur. J. Immunol.
Skin-resident memory CD8⁺ T cells trigger a state of tissue-wide pathogen 35, 383–390.
alert. Science 346, 101–105.
Dalmasso, A.P., Martinez, C., Sjodin, K., and Good, R.A. (1963). Studies on the
Bacchetta, R., Gambineri, E., and Roncarolo, M.G. (2007). Role of regulatory Role of the Thymus in Immunobiology; Reconstitution of Immunologic Capac-
T cells and FOXP3 in human diseases. J. Allergy Clin. Immunol. 120, 235, ity in Mice Thymectomized at Birth. J. Exp. Med. 118, 1089–1109.
quiz 236–237.
Dejaco, C., Duftner, C., Grubeck-Loebenstein, B., and Schirmer, M. (2006).
Bennett, C.L., Christie, J., Ramsdell, F., Brunkow, M.E., Ferguson, P.J., White- Imbalance of regulatory T cells in human autoimmune diseases. Immunology
sell, L., Kelly, T.E., Saulsbury, F.T., Chance, P.F., and Ochs, H.D. (2001). The 117, 289–300.
immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome
(IPEX) is caused by mutations of FOXP3. Nat. Genet. 27, 20–21. den Braber, I., Mugwagwa, T., Vrisekoop, N., Westera, L., Mögling, R., de
Boer, A.B., Willems, N., Schrijver, E.H., Spierenburg, G., Gaiser, K., et al.
Bergsbaken, T., and Bevan, M.J. (2015). Proinflammatory microenvironments (2012). Maintenance of peripheral naive T cells is sustained by thymus output
within the intestine regulate the differentiation of tissue-resident CD8⁺ T cells in mice but not humans. Immunity 36, 288–297.
responding to infection. Nat. Immunol. 16, 406–414.
DeWitt, W.S., Emerson, R.O., Lindau, P., Vignali, M., Snyder, T.M., Desmarais,
Beura, L.K., Hamilton, S.E., Bi, K., Schenkel, J.M., Odumade, O.A., Casey, C., Sanders, C., Utsugi, H., Warren, E.H., McElrath, J., et al. (2015). Dynamics
K.A., Thompson, E.A., Fraser, K.A., Rosato, P.C., Filali-Mouhim, A., et al. of the cytotoxic T cell response to a model of acute viral infection. J. Virol. 89,
(2016). Normalizing the environment recapitulates adult human immune traits 4517–4526.
in laboratory mice. Nature 532, 512–516.
Di Benedetto, S., Derhovanessian, E., Steinhagen-Thiessen, E., Goldeck, D.,
Bingaman, A.W., Patke, D.S., Mane, V.R., Ahmadzadeh, M., Ndejembi, M., Mu€ller, L., and Pawelec, G. (2015). Impact of age, sex and CMV-infection on
Bartlett, S.T., and Farber, D.L. (2005). Novel phenotypes and migratory prop- peripheral T cell phenotypes: results from the Berlin BASE-II Study. Bio-
erties distinguish memory CD4 T cell subsets in lymphoid and lung tissue. Eur. gerontology 16, 631–643.
J. Immunol. 35, 3173–3186.
Douek, D.C., McFarland, R.D., Keiser, P.H., Gage, E.A., Massey, J.M., Hay-
Blom, K., Braun, M., Ivarsson, M.A., Gonzalez, V.D., Falconer, K., Moll, M., nes, B.F., Polis, M.A., Haase, A.T., Feinberg, M.B., Sullivan, J.L., et al.
Ljunggren, H.G., Michae€lsson, J., and Sandberg, J.K. (2013). Temporal dy- (1998). Changes in thymic function with age and during the treatment of HIV
namics of the primary human T cell response to yellow fever virus 17D as it infection. Nature 396, 690–695.
matures from an effector- to a memory-type response. J. Immunol. 190,
2150–2158. Durek, P., Nordström, K., Gasparoni, G., Salhab, A., Kressler, C., de Almeida,
M., Bassler, K., Ulas, T., Schmidt, F., Xiong, J., et al.; DEEP Consortium (2016).
Booth, J.S., Toapanta, F.R., Salerno-Goncalves, R., Patil, S., Kader, H.A., Epigenomic Profiling of Human CD4+ T Cells Supports a Linear Differentiation
Safta, A.M., Czinn, S.J., Greenwald, B.D., and Sztein, M.B. (2014). Character- Model and Highlights Molecular Regulators of Memory Development. Immu-
ization and functional properties of gastric tissue-resident memory T cells from nity 45, 1148–1161.
children, adults, and the elderly. Front. Immunol. 5, 294.
Evans, C.J., Ho, Y., Daveson, B.A., Hall, S., Higginson, I.J., and Gao, W.;
Burt, T.D. (2013). Fetal regulatory T cells and peripheral immune tolerance in GUIDE_Care project (2014). Place and cause of death in centenarians: a pop-
utero: implications for development and disease. Am. J. Reprod. Immunol. ulation-based observational study in England, 2001 to 2010. PLoS Med. 11,
69, 346–358. e1001653.

Carpenter, D.J., Granot, T., Matsuoka, N., Senda, T., Kumar, B.V., Thome, Fernandez-Ruiz, D., Ng, W.Y., Holz, L.E., Ma, J.Z., Zaid, A., Wong, Y.C., Lau,
J.J.C., Gordon, C.L., Miron, M., Weiner, J., Connors, T., et al. (2018). Human L.S., Mollard, V., Cozijnsen, A., Collins, N., et al. (2016). Liver-Resident Mem-
immunology studies using organ donors: Impact of clinical variations on im- ory CD8+ T Cells Form a Front-Line Defense against Malaria Liver-Stage Infec-
mune parameters in tissues and circulation. Am. J. Transplant. 18, 74–88. Pub- tion. Immunity 45, 889–902.
lished online September 8, 2017.
Feuerer, M., Herrero, L., Cipolletta, D., Naaz, A., Wong, J., Nayer, A., Lee, J.,
Cheuk, S., Schlums, H., Gallais Sérézal, I., Martini, E., Chiang, S.C., Mar- Goldfine, A.B., Benoist, C., Shoelson, S., and Mathis, D. (2009). Lean, but
quardt, N., Gibbs, A., Detlofsson, E., Introini, A., Forkel, M., et al. (2017). not obese, fat is enriched for a unique population of regulatory T cells that
CD49a Expression Defines Tissue-Resident CD8+ T Cells Poised for Cytotoxic affect metabolic parameters. Nat. Med. 15, 930–939.
Function in Human Skin. Immunity 46, 287–300.
Fuertes Marraco, S.A., Soneson, C., Cagnon, L., Gannon, P.O., Allard, M.,
Chinn, I.K., Milner, J.D., Scheinberg, P., Douek, D.C., and Markert, M.L. (2013). Abed Maillard, S., Montandon, N., Rufer, N., Waldvogel, S., Delorenzi, M.,
Thymus transplantation restores the repertoires of forkhead box protein 3 and Speiser, D.E. (2015). Long-lasting stem cell-like memory CD8+ T cells
(FoxP3)+ and FoxP3- T cells in complete DiGeorge anomaly. Clin. Exp. Immu- with a naı̈ve-like profile upon yellow fever vaccination. Sci. Transl. Med. 7,
nol. 173, 140–149. 282ra48.

Cibrián, D., and Sánchez-Madrid, F. (2017). CD69: from activation marker to Ganusov, V.V., and De Boer, R.J. (2007). Do most lymphocytes in humans
metabolic gatekeeper. Eur. J. Immunol. 47, 946–953. really reside in the gut? Trends Immunol. 28, 514–518.

210 Immunity 48, February 20, 2018


Immunity

Review
Gattinoni, L., Lugli, E., Ji, Y., Pos, Z., Paulos, C.M., Quigley, M.F., Almeida, Jiang, X., Clark, R.A., Liu, L., Wagers, A.J., Fuhlbrigge, R.C., and Kupper, T.S.
J.R., Gostick, E., Yu, Z., Carpenito, C., et al. (2011). A human memory T cell (2012). Skin infection generates non-migratory memory CD8+ T(RM) cells
subset with stem cell-like properties. Nat. Med. 17, 1290–1297. providing global skin immunity. Nature 483, 227–231.

Gebhardt, T., Wakim, L.M., Eidsmo, L., Reading, P.C., Heath, W.R., and Car- Junge, S., Kloeckener-Gruissem, B., Zufferey, R., Keisker, A., Salgo, B., Fau-
bone, F.R. (2009). Memory T cells in nonlymphoid tissue that provide chere, J.C., Scherer, F., Shalaby, T., Grotzer, M., Siler, U., et al. (2007). Corre-
enhanced local immunity during infection with herpes simplex virus. Nat. lation between recent thymic emigrants and CD31+ (PECAM-1) CD4+ T cells in
Immunol. 10, 524–530. normal individuals during aging and in lymphopenic children. Eur. J. Immunol.
37, 3270–3280.
Gebhardt, T., Whitney, P.G., Zaid, A., Mackay, L.K., Brooks, A.G., Heath, W.R.,
Carbone, F.R., and Mueller, S.N. (2011). Different patterns of peripheral migra- Kaech, S.M., and Wherry, E.J. (2007). Heterogeneity and cell-fate decisions in
tion by memory CD4+ and CD8+ T cells. Nature 477, 216–219. effector and memory CD8+ T cell differentiation during viral infection. Immunity
27, 393–405.
Germain, R.N. (2002). T-cell development and the CD4-CD8 lineage decision.
Nat. Rev. Immunol. 2, 309–322. Klein, L., Kyewski, B., Allen, P.M., and Hogquist, K.A. (2014). Positive and
negative selection of the T cell repertoire: what thymocytes see (and don’t
Glennie, N.D., Yeramilli, V.A., Beiting, D.P., Volk, S.W., Weaver, C.T., and see). Nat. Rev. Immunol. 14, 377–391.
Scott, P. (2015). Skin-resident memory CD4+ T cells enhance protection
against Leishmania major infection. J. Exp. Med. 212, 1405–1414. Kohler, S., Bethke, N., Bothe, M., Sommerick, S., Frentsch, M., Romagnani,
C., Niedrig, M., and Thiel, A. (2012). The early cellular signatures of protective
Golding, A., Darko, S., Wylie, W.H., Douek, D.C., and Shevach, E.M. (2017). immunity induced by live viral vaccination. Eur J Immunol 42, 2363–2373.
Deep sequencing of the TCR-b repertoire of human forkhead box protein 3
(FoxP3)+ and FoxP3- T cells suggests that they are completely distinct and Kumar, B.V., Ma, W., Miron, M., Granot, T., Guyer, R.S., Carpenter, D.J.,
non-overlapping. Clin. Exp. Immunol. 188, 12–21. Senda, T., Sun, X., Ho, S.H., Lerner, H., et al. (2017). Human Tissue-Resident
Memory T Cells Are Defined by Core Transcriptional and Functional Signatures
Gordon, C.L., Miron, M., Thome, J.J., Matsuoka, N., Weiner, J., Rak, M.A., in Lymphoid and Mucosal Sites. Cell Rep. 20, 2921–2934.
Igarashi, S., Granot, T., Lerner, H., Goodrum, F., and Farber, D.L. (2017). Tis-
sue reservoirs of antiviral T cell immunity in persistent human CMV infection. Larbi, A., and Fulop, T. (2014). From ‘‘truly naı̈ve’’ to ‘‘exhausted senescent’’
J. Exp. Med. 214, 651–667. T cells: when markers predict functionality. Cytometry A 85, 25–35.
Goronzy, J.J., and Weyand, C.M. (2017). Successful and Maladaptive T Cell Li, B., Li, J., Devlin, B.H., and Markert, M.L. (2011). Thymic microenvironment
Aging. Immunity 46, 364–378. reconstitution after postnatal human thymus transplantation. Clin. Immunol.
140, 244–259.
Gourley, T.S., Wherry, E.J., Masopust, D., and Ahmed, R. (2004). Generation
and maintenance of immunological memory. Semin. Immunol. 16, 323–333. Mackay, L.K., Braun, A., Macleod, B.L., Collins, N., Tebartz, C., Bedoui, S.,
Carbone, F.R., and Gebhardt, T. (2015). Cutting edge: CD69 interference
Gurkan, S., Luan, Y., Dhillon, N., Allam, S.R., Montague, T., Bromberg, J.S.,
with sphingosine-1-phosphate receptor function regulates peripheral T cell
Ames, S., Lerner, S., Ebcioglu, Z., Nair, V., et al. (2010). Immune reconstitution
retention. J. Immunol. 194, 2059–2063.
following rabbit antithymocyte globulin. Am. J. Transplant. 10, 2132–2141.
Mackay, L.K., Minnich, M., Kragten, N.A., Liao, Y., Nota, B., Seillet, C., Zaid, A.,
Hammarlund, E., Lewis, M.W., Hansen, S.G., Strelow, L.I., Nelson, J.A.,
Man, K., Preston, S., Freestone, D., et al. (2016). Hobit and Blimp1 instruct a
Sexton, G.J., Hanifin, J.M., and Slifka, M.K. (2003). Duration of antiviral immu-
universal transcriptional program of tissue residency in lymphocytes. Science
nity after smallpox vaccination. Nat. Med. 9, 1131–1137.
352, 459–463.
Haynes, B.F., Martin, M.E., Kay, H.H., and Kurtzberg, J. (1988). Early events in
human T cell ontogeny. Phenotypic characterization and immunohistologic Mancebo, E., Clemente, J., Sanchez, J., Ruiz-Contreras, J., De Pablos, P.,
localization of T cell precursors in early human fetal tissues. J. Exp. Med. Cortezon, S., Romo, E., Paz-Artal, E., and Allende, L.M. (2008). Longitudinal
168, 1061–1080. analysis of immune function in the first 3 years of life in thymectomized neo-
nates during cardiac surgery. Clin. Exp. Immunol. 154, 375–383.
Haynes, B.F., Markert, M.L., Sempowski, G.D., Patel, D.D., and Hale, L.P.
(2000). The role of the thymus in immune reconstitution in aging, bone marrow Markert, M.L., Sarzotti, M., Ozaki, D.A., Sempowski, G.D., Rhein, M.E., Hale,
transplantation, and HIV-1 infection. Annu. Rev. Immunol. 18, 529–560. L.P., Le Deist, F., Alexieff, M.J., Li, J., Hauser, E.R., et al. (2003). Thymus trans-
plantation in complete DiGeorge syndrome: immunologic and safety evalua-
Hazenberg, M.D., Verschuren, M.C., Hamann, D., Miedema, F., and van Don- tions in 12 patients. Blood 102, 1121–1130.
gen, J.J. (2001). T cell receptor excision circles as markers for recent thymic
emigrants: basic aspects, technical approach, and guidelines for interpreta- Markert, M.L., Devlin, B.H., and McCarthy, E.A. (2010). Thymus transplanta-
tion. J. Mol. Med. (Berl.) 79, 631–640. tion. Clin. Immunol. 135, 236–246.

Hazenberg, M.D., Otto, S.A., de Pauw, E.S., Roelofs, H., Fibbe, W.E., Hamann, Masopust, D., Vezys, V., Marzo, A.L., and Lefrançois, L. (2001). Preferential
D., and Miedema, F. (2002). T-cell receptor excision circle and T-cell dynamics localization of effector memory cells in nonlymphoid tissue. Science 291,
after allogeneic stem cell transplantation are related to clinical events. Blood 2413–2417.
99, 3449–3453.
Masopust, D., Vezys, V., Usherwood, E.J., Cauley, L.S., Olson, S., Marzo, A.L.,
Hombrink, P., Helbig, C., Backer, R.A., Piet, B., Oja, A.E., Stark, R., Brasser, Ward, R.L., Woodland, D.L., and Lefrançois, L. (2004). Activated primary and
G., Jongejan, A., Jonkers, R.E., Nota, B., et al. (2016). Programs for the persis- memory CD8 T cells migrate to nonlymphoid tissues regardless of site of acti-
tence, vigilance and control of human CD8+ lung-resident memory T cells. Nat. vation or tissue of origin. J. Immunol. 172, 4875–4882.
Immunol. 17, 1467–1478.
McGovern, N., Shin, A., Low, G., Low, D., Duan, K., Yao, L.J., Msallam, R.,
Hondowicz, B.D., An, D., Schenkel, J.M., Kim, K.S., Steach, H.R., Krishna- Low, I., Shadan, N.B., Sumatoh, H.R., et al. (2017). Human fetal dendritic cells
murty, A.T., Keitany, G.J., Garza, E.N., Fraser, K.A., Moon, J.J., et al. (2016). promote prenatal T-cell immune suppression through arginase-2. Nature 546,
Interleukin-2-Dependent Allergen-Specific Tissue-Resident Memory Cells 662–666.
Drive Asthma. Immunity 44, 155–166.
€lsson, J., Mold, J.E., McCune, J.M., and Nixon, D.F. (2006). Regulation
Michae
Hori, S., Nomura, T., and Sakaguchi, S. (2003). Control of regulatory T cell of T cell responses in the developing human fetus. J. Immunol. 176,
development by the transcription factor Foxp3. Science 299, 1057–1061. 5741–5748.

Hudson, L.L., Louise Markert, M., Devlin, B.H., Haynes, B.F., and Sempowski, Miller, J.D., van der Most, R.G., Akondy, R.S., Glidewell, J.T., Albott, S., Maso-
G.D. (2007). Human T cell reconstitution in DiGeorge syndrome and HIV-1 pust, D., Murali-Krishna, K., Mahar, P.L., Edupuganti, S., Lalor, S., et al. (2008).
infection. Semin. Immunol. 19, 297–309. Human effector and memory CD8+ T cell responses to smallpox and yellow fe-
ver vaccines. Immunity 28, 710–722.
Jamieson, B.D., Douek, D.C., Killian, S., Hultin, L.E., Scripture-Adams, D.D.,
Giorgi, J.V., Marelli, D., Koup, R.A., and Zack, J.A. (1999). Generation of func- €lsson, J., Burt, T.D., Muench, M.O., Beckerman, K.P.,
Mold, J.E., Michae
tional thymocytes in the human adult. Immunity 10, 569–575. Busch, M.P., Lee, T.H., Nixon, D.F., and McCune, J.M. (2008). Maternal

Immunity 48, February 20, 2018 211


Immunity

Review
alloantigens promote the development of tolerogenic fetal regulatory T cells in Saule, P., Trauet, J., Dutriez, V., Lekeux, V., Dessaint, J.P., and Labalette, M.
utero. Science 322, 1562–1565. (2006). Accumulation of memory T cells from childhood to old age: central and
effector memory cells in CD4(+) versus effector memory and terminally differ-
Morawski, P.A., Qi, C.F., and Bolland, S. (2017). Non-pathogenic tissue-resi- entiated memory cells in CD8(+) compartment. Mech. Ageing Dev. 127,
dent CD8+ T cells uniquely accumulate in the brains of lupus-prone mice. 274–281.
Sci. Rep. 7, 40838.
Schenkel, J.M., and Masopust, D. (2014). Tissue-resident memory T cells. Im-
Moskowitz, D.M., Zhang, D.W., Hu, B., Le Saux, S., Yanes, R.E., Ye, Z., Buen- munity 41, 886–897.
rostro, J.D., Weyand, C.M., Greenleaf, W.J., and Goronzy, J.J. (2017). Epige-
nomics of human CD8 T cell differentiation and aging. Sci. Immunol. 2, Schenkel, J.M., Fraser, K.A., Beura, L.K., Pauken, K.E., Vezys, V., and Maso-
eaag0192. pust, D. (2014). T cell memory. Resident memory CD8 T cells trigger protective
innate and adaptive immune responses. Science 346, 98–101.
Mueller, S.N., and Mackay, L.K. (2016). Tissue-resident memory T cells: local
specialists in immune defence. Nat. Rev. Immunol. 16, 79–89. Seddiki, N., Santner-Nanan, B., Tangye, S.G., Alexander, S.I., Solomon, M.,
Lee, S., Nanan, R., and Fazekas de Saint Groth, B. (2006). Persistence of naive
€n, J.R., Westendorf, K., Fang, Z., Reinke, S., von Roth, P.,
Okhrimenko, A., Gru CD45RA+ regulatory T cells in adult life. Blood 107, 2830–2838.
€hl, A.A., Kudernatsch, R., Demski, S., et al. (2014). Human
Wassilew, G., Ku
memory T cells from the bone marrow are resting and maintain long-lasting Shin, H., and Iwasaki, A. (2012). A vaccine strategy that protects against gen-
systemic memory. Proc. Natl. Acad. Sci. USA 111, 9229–9234. ital herpes by establishing local memory T cells. Nature 491, 463–467.
Pallett, L.J., Davies, J., Colbeck, E.J., Robertson, F., Hansi, N., Easom, N.J.W., Shiow, L.R., Rosen, D.B., Brdicková, N., Xu, Y., An, J., Lanier, L.L., Cyster,
Burton, A.R., Stegmann, K.A., Schurich, A., Swadling, L., et al. (2017). IL-2high J.G., and Matloubian, M. (2006). CD69 acts downstream of interferon-alpha/
tissue-resident T cells in the human liver: Sentinels for hepatotropic infection. beta to inhibit S1P1 and lymphocyte egress from lymphoid organs. Nature
J. Exp. Med. 214, 1567–1580. 440, 540–544.
Panduro, M., Benoist, C., and Mathis, D. (2016). Tissue Tregs. Annu. Rev. Im- Silva, S.L., Albuquerque, A.S., Serra-Caetano, A., Foxall, R.B., Pires, A.R., Ma-
munol. 34, 609–633. toso, P., Fernandes, S.M., Ferreira, J., Cheynier, R., Victorino, R.M., et al.
(2016). Human naı̈ve regulatory T-cells feature high steady-state turnover
Peters, J.H., Koenen, H.J., Fasse, E., Tijssen, H.J., Ijzermans, J.N., Groenen, and are maintained by IL-7. Oncotarget 7, 12163–12175.
P.J., Schaap, N.P., Kwekkeboom, J., and Joosten, I. (2013). Human secondary
lymphoid organs typically contain polyclonally-activated proliferating regula- Silva, S.L., Albuquerque, A., Amaral, A.J., Li, Q.Z., Mota, C., Cheynier, R., Vic-
tory T cells. Blood 122, 2213–2223. torino, R.M.M., Pereira-Santos, M.C., and Sousa, A.E. (2017). Autoimmunity
and allergy control in adults submitted to complete thymectomy early in in-
€tter, A., Wu
Prelog, M., Keller, M., Geiger, R., Brandsta €rzner, R., Schweigmann,
fancy. PLoS ONE 12, e0180385.
U., Zlamy, M., Zimmerhackl, L.B., and Grubeck-Loebenstein, B. (2009). Thy-
mectomy in early childhood: significant alterations of the CD4(+)CD45RA(+) Skon, C.N., Lee, J.Y., Anderson, K.G., Masopust, D., Hogquist, K.A., and
CD62L(+) T cell compartment in later life. Clin. Immunol. 130, 123–132. Jameson, S.C. (2013). Transcriptional downregulation of S1pr1 is required
for the establishment of resident memory CD8+ T cells. Nat. Immunol. 14,
Purwar, R., Campbell, J., Murphy, G., Richards, W.G., Clark, R.A., and Kupper,
1285–1293.
T.S. (2011). Resident memory T cells (T(RM)) are abundant in human lung: di-
versity, function, and antigen specificity. PLoS ONE 6, e16245.
Soares, L.R., Tsavaler, L., Rivas, A., and Engleman, E.G. (1998). V7 (CD101)
Qi, Q., Liu, Y., Cheng, Y., Glanville, J., Zhang, D., Lee, J.Y., Olshen, R.A., ligation inhibits TCR/CD3-induced IL-2 production by blocking Ca2+ flux
Weyand, C.M., Boyd, S.D., and Goronzy, J.J. (2014). Diversity and clonal se- and nuclear factor of activated T cell nuclear translocation. J. Immunol. 161,
lection in the human T-cell repertoire. Proc. Natl. Acad. Sci. USA 111, 209–217.
13139–13144.
Steinert, E.M., Schenkel, J.M., Fraser, K.A., Beura, L.K., Manlove, L.S.,
Radenkovic, M., Uvebrant, K., Skog, O., Sarmiento, L., Avartsson, J., Storm, Igyártó, B.Z., Southern, P.J., and Masopust, D. (2015). Quantifying Memory
P., Vickman, P., Bertilsson, P.A., Fex, M., Korgsgren, O., and Cilio, C.M. CD8 T Cells Reveals Regionalization of Immunosurveillance. Cell 161,
(2017). Characterization of resident lymphocytes in human pancreatic islets. 737–749.
Clin. Exp. Immunol. 187, 418–427.
Stritesky, G.L., Jameson, S.C., and Hogquist, K.A. (2012). Selection of self-
Rosenberg, S.A. (2014). Decade in review-cancer immunotherapy: entering reactive T cells in the thymus. Annu. Rev. Immunol. 30, 95–114.
the mainstream of cancer treatment. Nat. Rev. Clin. Oncol. 11, 630–632.
Swaims-Kohlmeier, A., Haaland, R.E., Haddad, L.B., Sheth, A.N., Evans-
Sakaguchi, S., Takahashi, T., and Nishizuka, Y. (1982). Study on cellular events Strickfaden, T., Lupo, L.D., Cordes, S., Aguirre, A.J., Lupoli, K.A., Chen,
in postthymectomy autoimmune oophoritis in mice. I. Requirement of Lyt-1 C.Y., et al. (2016). Progesterone Levels Associate with a Novel Population of
effector cells for oocytes damage after adoptive transfer. J. Exp. Med. 156, CCR5+CD38+ CD4 T Cells Resident in the Genital Mucosa with Lymphoid
1565–1576. Trafficking Potential. J. Immunol. 197, 368–376.

Sakaguchi, S., Sakaguchi, N., Asano, M., Itoh, M., and Toda, M. (1995). Immu- Tanaskovic, S., Fernandez, S., Price, P., Lee, S., and French, M.A. (2010).
nologic self-tolerance maintained by activated T cells expressing IL-2 receptor CD31 (PECAM-1) is a marker of recent thymic emigrants among CD4+ T-cells,
alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance but not CD8+ T-cells or gammadelta T-cells, in HIV patients responding to
causes various autoimmune diseases. J. Immunol. 155, 1151–1164. ART. Immunol. Cell Biol. 88, 321–327.

Sakai, S., Kauffman, K.D., Schenkel, J.M., McBerry, C.C., Mayer-Barber, K.D., Teijaro, J.R., Turner, D., Pham, Q., Wherry, E.J., Lefrançois, L., and Farber,
Masopust, D., and Barber, D.L. (2014). Cutting edge: control of Mycobacte- D.L. (2011). Cutting edge: Tissue-retentive lung memory CD4 T cells mediate
rium tuberculosis infection by a subset of lung parenchyma-homing CD4 optimal protection to respiratory virus infection. J. Immunol. 187, 5510–5514.
T cells. J. Immunol. 192, 2965–2969.
Thiault, N., Darrigues, J., Adoue, V., Gros, M., Binet, B., Perals, C., Leobon, B.,
Sallusto, F., Lenig, D., Förster, R., Lipp, M., and Lanzavecchia, A. (1999). Two Fazilleau, N., Joffre, O.P., Robey, E.A., et al. (2015). Peripheral regulatory T
subsets of memory T lymphocytes with distinct homing potentials and effector lymphocytes recirculating to the thymus suppress the development of their
functions. Nature 401, 708–712. precursors. Nat. Immunol. 16, 628–634.

Sanchez Rodriguez, R., Pauli, M.L., Neuhaus, I.M., Yu, S.S., Arron, S.T., Harris, Thom, J.T., Weber, T.C., Walton, S.M., Torti, N., and Oxenius, A. (2015). The
H.W., Yang, S.H., Anthony, B.A., Sverdrup, F.M., Krow-Lucal, E., et al. (2014). Salivary Gland Acts as a Sink for Tissue-Resident Memory CD8(+) T Cells,
Memory regulatory T cells reside in human skin. J. Clin. Invest. 124, Facilitating Protection from Local Cytomegalovirus Infection. Cell Rep. 13,
1027–1036. 1125–1136.

Sathaliyawala, T., Kubota, M., Yudanin, N., Turner, D., Camp, P., Thome, J.J., Thome, J.J.C., Yudanin, N., Ohmura, Y., Kubota, M., Grinshpun, B., Sathaliya-
Bickham, K.L., Lerner, H., Goldstein, M., Sykes, M., et al. (2013). Distribution wala, T., Kato, T., Lerner, H., Shen, Y., and Farber, D.L. (2014). Spatial map of
and compartmentalization of human circulating and tissue-resident memory human T cell compartmentalization and maintenance over decades of life. Cell
T cell subsets. Immunity 38, 187–197. 159, 814–828.

212 Immunity 48, February 20, 2018


Immunity

Review
Thome, J.J., Bickham, K.L., Ohmura, Y., Kubota, M., Matsuoka, N., Gordon, polarized CX3CR1+ cytotoxic CD4+ T cells associated with protective immu-
C., Granot, T., Griesemer, A., Lerner, H., Kato, T., and Farber, D.L. (2016a). nity. Proc. Natl. Acad. Sci. USA 112, E4256–E4263.
Early-life compartmentalization of human T cell differentiation and regulatory
function in mucosal and lymphoid tissues. Nat. Med. 22, 72–77. Wells, W.J., Parkman, R., Smogorzewska, E., and Barr, M. (1998). Neonatal
thymectomy: does it affect immune function? J. Thorac. Cardiovasc. Surg.
Thome, J.J., Grinshpun, B., Kumar, B.V., Kubota, M., Ohmura, Y., Lerner, H., 115, 1041–1046.
Sempowski, G.D., Shen, Y., and Farber, D.L. (2016b). Longterm maintenance
of human naive T cells through in situ homeostasis in lymphoid tissue sites. Sci.
Wieten, R.W., Goorhuis, A., Jonker, E.F.F., de Bree, G.J., de Visser, A.W., van
Immunol. 1, aah6506.
Genderen, P.J.J., Remmerswaal, E.B.M., Ten Berge, I.J.M., Visser, L.G., Gro-
Trimble, C.L., Clark, R.A., Thoburn, C., Hanson, N.C., Tassello, J., Frosina, D., busch, M.P., and van Leeuwen, E.M.M. (2016a). 17D yellow fever vaccine
Kos, F., Teague, J., Jiang, Y., Barat, N.C., and Jungbluth, A.A. (2010). Human elicits comparable long-term immune responses in healthy individuals and im-
papillomavirus 16-associated cervical intraepithelial neoplasia in humans ex- mune-compromised patients. J. Infect. 72, 713–722.
cludes CD8 T cells from dysplastic epithelium. J. Immunol. 185, 7107–7114.
Wieten, R.W., Jonker, E.F., van Leeuwen, E.M., Remmerswaal, E.B., Ten
Turner, D.L., Bickham, K.L., Thome, J.J., Kim, C.Y., D’Ovidio, F., Wherry, E.J., Berge, I.J., de Visser, A.W., van Genderen, P.J., Goorhuis, A., Visser, L.G.,
and Farber, D.L. (2014). Lung niches for the generation and maintenance of tis- Grobusch, M.P., and de Bree, G.J. (2016b). A Single 17D Yellow Fever Vacci-
sue-resident memory T cells. Mucosal Immunol. 7, 501–510. nation Provides Lifelong Immunity; Characterization of Yellow-Fever-Specific
Neutralizing Antibody and T-Cell Responses after Vaccination. PLoS ONE 11,
Ugur, M., Schulz, O., Menon, M.B., Krueger, A., and Pabst, O. (2014). Resident e0149871.
CD4+ T cells accumulate in lymphoid organs after prolonged antigen expo-
sure. Nat. Commun. 5, 4821. Willinger, T., Freeman, T., Hasegawa, H., McMichael, A.J., and Callan, M.F.
(2005). Molecular signatures distinguish human central memory from effector
van den Broek, T., Delemarre, E.M., Janssen, W.J., Nievelstein, R.A., Broen, memory CD8 T cell subsets. J. Immunol. 175, 5895–5903.
J.C., Tesselaar, K., Borghans, J.A., Nieuwenhuis, E.E., Prakken, B.J., Mokry,
M., et al. (2016). Neonatal thymectomy reveals differentiation and plasticity
within human naive T cells. J. Clin. Invest. 126, 1126–1136. Wong, M.T., Ong, D.E., Lim, F.S., Teng, K.W., McGovern, N., Narayanan, S.,
Ho, W.Q., Cerny, D., Tan, H.K., Anicete, R., et al. (2016). A High-Dimensional
Vieira Braga, F.A., Hertoghs, K.M., Kragten, N.A., Doody, G.M., Barnes, N.A., Atlas of Human T Cell Diversity Reveals Tissue-Specific Trafficking and Cyto-
Remmerswaal, E.B., Hsiao, C.C., Moerland, P.D., Wouters, D., Derks, I.A., kine Signatures. Immunity 45, 442–456.
et al. (2015). Blimp-1 homolog Hobit identifies effector-type lymphocytes in
humans. Eur. J. Immunol. 45, 2945–2958. Woon, H.G., Braun, A., Li, J., Smith, C., Edwards, J., Sierro, F., Feng, C.G.,
Khanna, R., Elliot, M., Bell, A., et al. (2016). Compartmentalization of Total
Vrisekoop, N., den Braber, I., de Boer, A.B., Ruiter, A.F., Ackermans, M.T., van and Virus-Specific Tissue-Resident Memory CD8+ T Cells in Human Lymphoid
der Crabben, S.N., Schrijver, E.H., Spierenburg, G., Sauerwein, H.P., Hazen- Organs. PLoS Pathog. 12, e1005799.
berg, M.D., et al. (2008). Sparse production but preferential incorporation of
recently produced naive T cells in the human peripheral pool. Proc. Natl. Zens, K.D., Chen, J.-K., and Farber, D.L. (2016). Vaccine-generated lung tis-
Acad. Sci. USA 105, 6115–6120. sue-resident memory T cells provide heterosubtypic protection to influenza
infection. JCI Insight 1, e85832.
Watanabe, N., Wang, Y.H., Lee, H.K., Ito, T., Wang, Y.H., Cao, W., and Liu, Y.J.
(2005). Hassall’s corpuscles instruct dendritic cells to induce CD4+CD25+ reg-
Zhu, J., Koelle, D.M., Cao, J., Vazquez, J., Huang, M.L., Hladik, F., Wald, A.,
ulatory T cells in human thymus. Nature 436, 1181–1185.
and Corey, L. (2007). Virus-specific CD8+ T cells accumulate near sensory
Watanabe, R., Gehad, A., Yang, C., Scott, L.L., Teague, J.E., Schlapbach, C., nerve endings in genital skin during subclinical HSV-2 reactivation. J. Exp.
Elco, C.P., Huang, V., Matos, T.R., Kupper, T.S., and Clark, R.A. (2015). Human Med. 204, 595–603.
skin is protected by four functionally and phenotypically discrete populations
of resident and recirculating memory T cells. Sci. Transl. Med. 7, 279ra39. Zhu, J., Peng, T., Johnston, C., Phasouk, K., Kask, A.S., Klock, A., Jin, L.,
Diem, K., Koelle, D.M., Wald, A., et al. (2013). Immune surveillance by
Weiskopf, D., Bangs, D.J., Sidney, J., Kolla, R.V., De Silva, A.D., de Silva, A.M., CD8aa+ skin-resident T cells in human herpes virus infection. Nature 497,
Crotty, S., Peters, B., and Sette, A. (2015). Dengue virus infection elicits highly 494–497.

Immunity 48, February 20, 2018 213

You might also like