Anda di halaman 1dari 11

REVIEWS

FOXP3+ regulatory T cells in the


human immune system
Shimon Sakaguchi*, Makoto Miyara, Cristina M. Costantino and David A. Hafler||

Abstract | Forkhead box P3 (FOXP3)+ regulatory T (TReg) cells are potent mediators of dominant self
tolerance in the periphery. But confusion as to the identity, stability and suppressive function of
human TReg cells has, to date, impeded the general therapeutic use of these cells. Recent studies
have suggested that human TReg cells are functionally and phenotypically diverse. Here we
discuss recent findings regarding human TReg cells, including the ontogeny and development of
TReg cell subsets that have naive or memory phenotypes, the unique mechanisms of suppression
mediated by TReg cell subsets and factors that regulate TReg cell lineage commitment. We
discuss future studies that are needed for the successful therapeutic use of human TReg cells.

*Department of Experimental
Pathology, Institute for
Frontier Medical Sciences,
Kyoto University, Kyoto
6068507, Japan, and WPI
Immunology Frontier
Research Center, Osaka
University, Suita 5650871,
Japan.

Internal Medicine
Department 2, Laboratoire
dimmunochimie and
Laboratoire dimmunologie
cellulaire et tissulaire
(INSERM UMRS945), APHP,
Hpital PitiSalptrire,
75013 Paris, France.

Department of
Pharmacology and Systems
Therapeutics, Mount Sinai
School of Medicine, New York,
New York 10029, USA.
||
Harvard Medical School,
Boston, Massachusetts
02115, USA, and Department
of Neurology and
Immunobiology, Yale School
of Medicine, New Haven,
Connecticut 06520, USA.
Correspondence to S.S. and
D.A.H.
emails: shimon@frontier.
kyotou.ac.jp;
hafler@broad.mit.edu
All authors contributed
equally to this article.
doi:10.1038/nri2785
Published online 18 June 2010

Regulatory T (TReg) cells expressing the transcription


factor forkhead box P3 (FOXP3) are naturally present
in the immune system. They are indispensable for the
maintenance of dominant self tolerance and immune
homeostasis. Their dysfunction (for example, owing to
FOXP3 gene mutation) causes fatal autoimmune disease, immunopathology and allergy 1. FOXP3+ TReg cells,
most of which are CD4+ T cells that express CD25 (the
interleukin-2 (IL-2) receptor -chain), can suppress the
activation, proliferation and effector functions such
as cytokine production of a wide range of immune
cells, including CD4+ and CD8+ T cells, natural killer
(NK) and NKT cells, B cells and antigen-presenting cells
(APCs) in vitro and in vivo1. This unique ability to control immune responses makes FOXP3+ TReg cells central
in the prevention of autoimmune disease, immunopathology and allergy, as well as in the maintenance of
allograft tolerance and fetalmaternal tolerance during
pregnancy 2. As a double-edged sword, FOXP3+ TReg cells
can also suppress antitumour immune responses and
favour tumour progression. Thus, despite the unsuccessful attempts by immunologists in the last quarter of the
past century to define TReg cells at the molecular level3,4,
TReg cell-related immunobiology and molecular biology
have now become a field of intense investigation, with
FOXP3+ TReg cells as a primary target in the search for
new clinical applications5.
In addition to CD4 +CD25 +FOXP3 + TReg cells, it
has been shown that other T cells also possess regulatory activity and can, at least in certain conditions,
prevent autoimmunity in rodents6. Most of these cells
such as IL-10-secreting TR1 cells, transforming growth
factor- (TGF)-secreting T helper 3 (TH3) cells and

certain CD4CD8 T cells and CD8+CD28 T cells


are adaptively regulatory: that is, they acquire regulatory functions following specific antigenic stimulation
in particular cytokine milieus. They therefore contrast
with naturally occurring CD4+FOXP3+ TReg cells, most
of which are developmentally determined in the thymus
as a distinct T cell subpopulation that is specialized for
suppressive function.
Human TReg cells were first characterized as CD4+CD25+
T cells in 2001 by several groups712 based on the finding
in 1995 that mouse TReg cells constitutively express CD25
(Ref. 13). Similarly, in 2003, Foxp3 was described as a master control gene for mouse TReg cell development and function1416, and subsequent studies have confirmed FOXP3
as a specific marker for human TReg cells17 (Timeline).
However, it was then shown that both CD25 and FOXP3
expression could be induced in human naive CD4+ T cells
through cell activation, obscuring the identification of
FOXP3+ T cells as pure TReg cells18. As a result, some confusion has prevailed as to the phenotype, the function and
the stability of FOXP3+ TReg cells in humans, producing
discrepancies in the literature regarding the phenotypic
and functional characterization of TReg cells in healthy
individuals and patients with immunological diseases.
Recent studies have shown that human CD4+FOXP3+
T cells are not homogeneous in gene expression, phenotype and suppressive function, and indicated that a
new basis for reliable delineation of human TReg cells is
required5,19,20. In this Review, we discuss the development
of human TReg cells, the definition of TReg cell subsets based
on the expression of FOXP3, CD45RA and CD45RO, the
phenotypes and functions of FOXP3+ TReg cell subsets and
their clinical applications.

490 | juLy 2010 | VOLuMe 10

www.nature.com/reviews/immunol
2010 Macmillan Publishers Limited. All rights reserved

REVIEWS
Timeline | TReg cell discovery
Suppression mediated
by thymus-derived
cells116

1969

1970

Day 3
thymectomy120

CD45RBlow as a
surface marker
for TReg cells118

1985

CD5hi as a
surface
marker for
TReg cells117

1990

In vitro
suppression
assays69,119

1995

CD25 as a
surface marker
for TReg cells13

1998

Identification of
CD4+CD25+/hi
cells as TReg cells
in humans712

2000

Dysfunctional
FOXP3 is
responsible
for IPEX3941

2001

Identification of naive TReg cells6466


FOXP3-specific monoclonal
antibodies available for flow
cytometry17

2003

2005

FOXP3 as a master
control gene for TReg cell
development and
function1416
Induction of FOXP3 in
activated CD4+ T cells49

2006

Identification of
ICOS+ TReg cells20

2008

CD127low as a marker
for CD4+FOXP3+
T cells59,60

2009

Functional delineation
and differentiation
dynamics of FOXP3+
T cell subsets5

Identification of
HLA-DR+ TReg cells19

Blue, mouse system; red, human system. FOXP3, forkhead box P3; ICOS, inducible T cell co-stimulator; IPEX, immunodysregulation,
polyendocrinopathy and enteropathy, X-linked syndrome; TReg, regulatory T.

Dominant self tolerance


Refers to the active suppression
of an autoimmune response
in vitro or in vivo by suppressor
cells, including regulatory
T cells. By contrast, deletional
tolerance and anergy are
referred to as recessive or
passive self tolerance
mechanisms. Dominant self
tolerance is transferable to
naive recipients, whereas
recessive self tolerance is not.

Central tolerance
Self tolerance that is created at
the level of the central lymphoid
organs. Developing T cells in the
thymus, and B cells in the bone
marrow, that strongly recognize
self antigen face deletion or
anergy induction.

Self tolerance
Tolerance to an individuals own
tissue antigens that is achieved
through both central and
peripheral tolerance
mechanisms, including T cell
deletion, anergy and immune
regulation. Without both central
and peripheral tolerance
mechanisms the immune system
would be unable to distinguish
self from foreign antigen,
resulting in autoimmunity.

Hassalls corpuscles
Small clusters or concentric
whorls of stratified keratinizing
epithelium in the thymic
medulla. They are probably
end-stage differentiated
epithelial cells that participate
in negative selection of
thymocytes and/or that
undergo apoptosis themselves.

Development of human TReg cells


Thymic origin of natural TReg cells. Before the discovery and characterization of naturally occurring TReg
cells, the cause of spontaneous autoimmune disease was
largely attributed to aberrant central tolerance. The role
of TReg cells in self tolerance was implicated by two animal models of autoimmunity: autoimmune disease was
induced in mice by neonatal thymectomy on day 24
after birth and in rats by adult thymectomy and subsequent rounds of X-ray irradiation3. The autoimmune
disease induced by either method can be prevented by
the transfer of CD4+ T cells from normal mice or rats.
These findings suggest that the normal thymus produces T cells with an autoimmune-preventive function,
that their ontogeny in mice can be disrupted by neonatal thymectomy and that they are highly proliferative
and hence radio-sensitive.
Direct evidence of the thymic origin of TReg cells and
their persistence in the periphery was shown by the
adoptive transfer of thymocyte or peripheral T cell suspensions depleted of CD4+CD25+ T cells, which results
in the development of various autoimmune diseases in
syngeneic T cell-deficient mice3,13,21. Ontogenically,
CD4 +CD25 + T cells (and FOXP3 + T cells, as later
shown22) become detectable in the periphery ~3 days
after birth, indicating that neonatal thymectomy abrogates thymic production of TReg cells3,23. Furthermore,
depletion of peripheral FOXP3+ T cells for a limited
time period results in autoimmunity in mice23,24. Based
on these findings, thymus-derived CD4+FOXP3+ T cells
are thought to be natural TReg cells.
Although immature FOXP3+ thymocytes have been
identified in the human thymus2528, little is known
about the requirements for thymic TReg cell development in humans. There are, however, several conditions
required for natural TReg cell development in mice1;
thymic stromal cells, including cortical and medullary thymic epithelial cells and dendritic cells (DCs),
contribute to TReg cell differentiation and selection,

although it is controversial how and to what extent each


stromal cell component is required for this process1,29.
Thymic development of TReg cells requires high-affinity
interactions between their T cell receptor (TCR) and
self-peptideMHC complexes presented by thymic
stromal cells30. These cells also provide co-stimulatory
signals that are necessary for TReg cell development as
shown by the decrease in the number of TReg cells generated in the thymus following loss of CD40 or CD28
expression1. Furthermore, both IL-2 and IL-7 in the
thymic microenvironment are required for TReg cell
development in mice31. Given the known similarities
between mouse and human thymocyte development32,
it is likely that many of these requirements for TReg cells
in mice are similar for human TReg cell development.
However, a unique structure in humans Hassalls
corpuscles in the thymic medulla may contribute
to forming a specialized microenvironment required to
support nascent TReg cells33. Hassalls corpuscles secrete
thymic stromal lymphopoietin (TSLP), which activates
immature, migratory CD11c+ DCs in the thymus and
upregulates their expression of co-stimulatory molecules33,34. TSLP-activated DCs seem to induce FOXP3
expression in immature CD4+CD8CD25 thymocytes,
but not in mature naive CD4+ T cells derived from
peripheral blood 35. TSLP-activated DCs may contribute to the positive selection of high-affinity selfreactive thymocytes and support their differentiation
into mature FOXP3+ TReg cells36.
TReg cell development in humans and mice may follow a similar course of ontogeny, although in humans
most of T cell developmental events take place in utero.
The human thymus produces mature T cells as early
as the thirteenth week of gestation37, and by the fourteenth week, ~7% of mature CD4+ thymocytes express
high levels of CD25, a phenotypic marker associated
with TReg cell function in adult peripheral blood38. At
this stage of development, the fetal organs are reportedly colonized by CD4+CD25hi TReg cells. These fetal

NATuRe ReVIewS | Immunology

VOLuMe 10 | juLy 2010 | 491


2010 Macmillan Publishers Limited. All rights reserved

REVIEWS
200223
Zinc-finger
domain

1193
Proline-rich region

240261
Leucine-zipper
domain

338421
Forkhead
domain

Homodimerization

Interaction
with DNA

Domains

FOXP3A
Exon 2
70105

FOXP3B

L-X-X-L-L motif
Interaction
with ROR
Important for inhibiting
NFAT-mediated transcription

Function

Interaction with
NF-B and NFAT

106190
Interaction with HDAC
No FOXP3

Low FOXP3

CTLA4CD28low
CD95CD25+/IL-2+
Non-suppressive

High FOXP3

CTLA4+CD28hi
CD95+CD25+IL-2+/
Non-suppressive

CTLA4hiCD28hi
CD95+CD25++IL-2
Suppressive

Figure 1 | FoXP3 expression in human TReg cells. Schematic of forkhead box P3,
isoform A (FOXP3A) and FOXP3B protein structure, with known domains listed above
Nature Reviews | Immunology
and putative function below. Expression of FOXP3 at low levels induces a limited TReg cell
phenotype but not suppressive function, which is only gained when FOXP3 is highly
expressed. CTLA4, cytotoxic T lymphocyte antigen 4; HDAC, histone deacetylase;
IL, interleukin; NF-B, nuclear factor-B; NFAT, nuclear factor of activated T cells;
ROR, retinoic acid receptor-related orphan receptor-.

thymus-derived TReg cells express FOXP3 and are


functionally suppressive when co-cultured with effector T cells38. These data indicate that functional human
TReg cells begin to develop in utero.

Scurfy mice
mice with a spontaneous
mutation in Foxp3, which
leads to a rapidly fatal lymphoproliferative disease, causing
death by ~4 weeks of age.

Immunodysregulation,
polyendocrinopathy,
enteropathy, Xlinked (IPEX)
syndrome
A disease caused by mutations
in FOXP3 and characterized by
refractory enteritis,
autoimmune endocrinopathies,
including type 1 diabetes,
thyroiditis and allergy.

The transcription factor FOXP3 in humans and mice.


The key role of FOXP3 gene in the maintenance of
self tolerance was first shown in scurfy mice and subsequently in patients with immunodysregulation, polyendocrinopathy, enteropathy, X-linked (iPeX) syndrome as the
causative genetic anomaly that results in severe autoimmune diseases and allergy, which resemble the diseases
observed following depletion of CD4+CD25+ TReg cells in
rodents3941. Importantly, ectopic expression of FOXP3
in naive mouse CD4+ T cells confers suppressive activity
and induces the expression of TReg cell-associated signature molecules such as CD25, cytotoxic T lymphocyte
antigen 4 (CTLA4) and glucocorticoid-induced TNFreceptor-related protein (GITR)14,16,24. expression of
these receptors also correlates with FOXP3 expression
in human CD4+ T cells42. This induction of suppressive activity in conventional T cells by ectopic FOXP3
expression, together with the development of autoimmune disease in FOXP3 mutant or deficient mice,
indicates that FOXP3 is a master regulator for TReg cell
differentiation and function43.
Humans express two main isoforms of FOXP3, either
of which can confer regulatory function when strongly
overexpressed42,44,45. The main deletional isoform of
FOXP3 (FOXP3B) lacks the proline-rich exon 2, which

encodes the Leu-X-X-Leu-Leu motif that is required


for binding to the transcription factor retinoic acid
receptor-related orphan receptor- (ROR)46, and lacks
amino-terminal residues that may mediate the interaction
with nuclear factor of activated T cells (NFAT) and the
resulting transcriptional repression47,48 (fiG. 1). The role of
FOXP3B in human TReg cell biology remains unclear.
Although the transient expression of FOXP3 does
not enable suppression, sustained FOXP3 expression by
activated T cells can confer regulatory competence44,4951
(fiG. 1). Furthermore, in human CD4+ TReg cells, stable
and high FOXP3 expression is required for suppressive function, and loss of FOXP3 expression over time
owing to long-term culture decreases the ability of
formerly potent TReg cells to suppress52,53.
In contrast to the human system, mouse naive CD4+
T cells lack transient and promiscuous expression of
FOXP3 following activation. However, conventional
mouse T cells readily convert to FOXP3+ TReg cells following in vitro stimulation with TGF or retinoic acid54.
Such induced or adaptive TReg cells express many of
the phenotypic markers associated with regulatory
function, such as FOXP3, CD25 and CTLA4, but they
do not exhibit the signature gene transcription profile of
ex vivo natural CD4+CD25+FOXP3+ TReg cells described
in Ref. 55. Indeed, much of the TGF-induced regulatory profile is FOXP3 independent, as it is similarly
expressed in TGF-treated CD4+ T cells derived from
FOXP3-deficient scurfy mice56.
In humans, in contrast with naturally occurring
CD4+FOXP3+ T cells, FOXP3+ T cells induced from
naive CD4+ T cells by in vitro TCR stimulation and
TGF show little in vitro suppressive activity, but
they can secrete pro-inflammatory cytokines50,57. This
indicates that conventional CD4+ T cells may require
additional factors, in addition to the functional and
phenotypic changes induced by FOXP3 expression,
to acquire full suppressive activity in humans. It
remains to be determined whether induced FOXP3+
TReg cells are physiologically present in vivo as functionally competent suppressive cells in humans and,
if this is the case, to what extent peripheral FOXP3+
TReg cells are converted from naive T cells or derived
from the thymus.
As mentioned above, human FOXP3 is expressed by
both activated and regulatory T cells in the peripheral
blood, as are other known TReg cell biomarkers such as
CD25, CTLA4, GITR and CD95 (also known as FAS)
(TABle 1). This upregulation of FOXP3 in activated T cells
could be one component of the homeostatic programme
initiated by these cells to exert negative feedback during
the course of an immune response. Importantly, it also
shows that FOXP3+ T cells in humans are functionally
heterogeneous, as discussed below.

Delineation of human TReg cells


As detection of intracellular FOXP3 makes it difficult
to conduct a functional assessment of TReg cells, cell
surface markers are crucial for identifying viable TReg
cells. In mice, CD4+FOXP3+ TReg cells in naive mice
can be identified by the expression CD25 (Ref. 13) .

492 | juLy 2010 | VOLuMe 10

www.nature.com/reviews/immunol
2010 Macmillan Publishers Limited. All rights reserved

REVIEWS
Table 1 | TReg cell markers
Transcription Activation
factor
and memory

Homing and
origin

Suppressive
and effector
function

Apoptosis,
survival or
other

FOXP3

CD62L
CCR4
CCR6
CCR9
CD103
CD304
CD31
Lack of CD49d

CTLA4
ICOS
CD39CD73
LAP
Granzyme B
Galectin 1
Galectin 10
TRANCE
CD80 and CD86
IL-10
IL-17
CD2
Lack of IL-2

CD27
OX40
CD95
PD1
GITR
Galectin 3
GARP
MS4A4B
IL-1R
CD6

CD45RA
CD45RO
CD25
HLA-DR
Lack of CD127
CD69

CCR, CC-chemokine receptor; CTLA4, cytotoxic T lymphocyte antigen 4; FOXP3, forkhead


box P3; GARP, glycoprotein A repetitions predominant; GITR, glucocorticoid-induced
TNF-receptor-related protein; ICOS, inducible T cell co-stimulator; IL, interleukin; LAP,
latency-associated peptide; MS4A4B, membrane-spanning 4-domains, subfamily A, member 4B;
PD1, programmed cell death 1; R, receptor; TRANCE, TNF-related activation-induced
cytokine; TReg, regulatory T.

Anergy
A state of non-responsiveness
to antigen. Anergic B or T cells
cannot respond to their
cognate antigens under
optimal conditions of
stimulation.

However, CD25 expression cannot be used in human


studies as peripheral blood isolated from an outbred
human population contains up to 30% CD4+CD25+
T cells. Only ~12% of cells with the highest CD25
expression have been shown to be functionally suppressive and can be considered TReg cells7,58. Furthermore,
given that the expression levels of FOXP3 and CD25
are proportional in human FOXP3+ T cells5, isolation
of T cells expressing high levels of CD25 excludes
FOXP3lowCD25mid TReg cells, which have a naive phenotype (CD45RAhiCD45RO) (see below). In addition,
for practical purposes, there is no firm criterion as to
where the boundary lies between intermediate and
high CD25 expression. This fuzzy boundary has hindered the reproducibility of clinical data analysing the
number and function of TReg cells, particularly under
inflammatory conditions in which activated T cells
express CD25.
Recently, several groups showed that the lack of cell
surface CD127 (also known as IL-7 receptor -chain)
can be a useful alternative to CD25 for the delineation
and purification of human TReg cells: FOXP3 expression
and suppressive ability are enriched in CD4+ T cells that
express low levels of CD127 (RefS 59,60). However, conventional CD4+ T cells tend to downregulate CD127
expression after activation61. Therefore CD127 expression alone cannot accurately discriminate TReg cells
from activated T cells ex vivo 62. Alternatively, CD62L
(also known as L-selectin) expression, although not
restricted to TReg cells, may be used to differentiate
between TReg cells, which are CD25hiCD127lowCD62L+,
and recently activated conventional CD4+ T cells, which
are CD62Llow (Ref. 63).
In addition, the combined use of CD127 and CD25
to isolate pure TReg cells is compromised by the existence of a non-regulatory FOXP3-expressing CD4 +
T cell population in the periphery in humans. Indeed
this population, which expresses CD45RO and low
levels of FOXP3, has low to intermediate CD127

expression but does not suppress effector cells in vitro


and produces pro-inflammatory cytokines, including
IL-2, interferon- (IFN) and IL-17 (Ref. 5). These nonregulatory FOXP3low T cells might be activated CD4+
effector T cells that have been shown in vitro to upregulate the expression of FOXP3 without the acquisition
of a suppressive ability. The difference between these
non-regulatory FOXP3+ T cells and functional FOXP3+
TReg cells may be linked to the methylation status of
the FOXP3 gene, which is incompletely demethylated
in CD45RO+FOXP3low non-regulatory T cells but is
completely demethylated in FOXP3+ TReg cells with
suppressive activity 5. The existence of this non-regulatory FOXP3low T cell population in normal individuals
precludes the use of the FOXP3 expression as a sole
marker for human TReg cells.
Heterogeneity of human TReg cells. As suggested above,
human FOXP3+ T cells are phenotypically and functionally heterogeneous, including suppressive and nonsuppressive T cells. Recent studies have shown that the
expression of CD45RA or CD45RO, which is mutually
exclusive in FOXP3+ as well as other T cells, is a particularly useful marker when combined with CD25 and/or
FOXP3 expression5,6466.
CD45RO +CD25 hiCD4 + TReg cells are similar to
mouse TReg cells in terms of the expression of CD25
and other TReg cell markers, potent suppressive activity, and apparent anergic state, and were therefore originally thought to be the human counterpart of mouse
CD25+CD4+ TReg cells712. However, it has now become
clear that, unlike in mice, human FOXP3+CD4+ T cells
with a naive phenotype (that is, expressing CD45RA
but not CD45RO) also possess potent suppressive functions and are present in peripheral blood and prevalent
in cord blood6466. A comparison of gene expression
as well as in vitro and in vivo behaviour between
CD45RA+FOXP3+ and CD45RO+FOXP3+ TReg cells has
revealed that these subsets are functionally different but
developmentally related5 (fiG. 2).
CD45RA + FOXP3 low naive T Reg cells. Naive TReg
cells are characterized by their surface expression of
CD45RA and their low levels of intranuclear FOXP3
(RefS 5,20,66). expression of CD45RA without concomitant expression of CD45RO is a phenotypic marker for
naive T cells that have not experienced TCR stimulation-mediated maturation. However, based on mouse
data, TReg cells need to be stimulated continuously by
their cognate antigen, presumably self antigen, to be
maintained in the periphery 67. Therefore, although they
express CD45RA, these TReg cells cannot be considered
strictly naive T cells. In fact, it has been known for more
than 20 years that activated CD4+ T cells can express
CD45RA68. However, most CD45RA+FOXP3low naive
TReg cells also express CD31 (also known as PeCAM1),
a cell surface marker specific for recent thymic emigrants, whereas virtually all CD45RO + TReg cells do
not. Therefore, it can be concluded that most, if not
all, thymus-derived TReg cells found in the periphery are
CD45RA+FOXP3low naive TReg cells5,20,66. The absence

NATuRe ReVIewS | Immunology

VOLuMe 10 | juLy 2010 | 493


2010 Macmillan Publishers Limited. All rights reserved

REVIEWS
Thymus

FOXP3
CD45RA+ CD45RO
CD127+ CD25 HLA-DR
CD95 CTLA4

FOXP3low
CD45RA+ CD45RO
CD127low/ CD25hi HLA-DR
CD95 CTLA4
Naive
TReg cell

Naive
T cell

FOXP3low
CD45RA CD45RO+
CD127low/ CD25hi HLA-DR+
CD95+ CTLA4+ CD69+ IL-2+
ICOS+/

FOXP3
CD45RA CD45RO+
CD127+ CD25+ CD62L+
CD95+ CTLA4+

?
Activated
T cell

Activated converted
TReg-like cell
FOXP3+
CD45RA CD45RO+
CD127+/ CD25+ CD62L
CD95+ CTLA4+
IL-17+ and/or IL-10+
ICOS+/

Memory
T cell

Effector
TReg cell

FOXP3hi
CD45RA CD45RO+
CD127low CD25hi HLA-DR
CD95+ CTLA4+ Ki67+
ICOS+/

Terminal
effector
TReg cell

FOXP3hi
CD45RA CD45RO+
CD127 CD25hi HLA-DR+
CD95+ CTLA4+ Ki67+
ICOS+/

Figure 2 | TReg cell differentiation. Phenotypic markers are indicated during CD4+ T cell differentiation into the
Nature
| Immunology
conventional T cell and regulatory T (TReg) cell lineages. All T cell lineages originate in the thymus
andReviews
emigrate
as naive
CD45RA+ T cells. Activation of naive T cells in the periphery induces their differentiation into both conventional and
regulatory subsets. Conventional T cells can further differentiate into memory T cells, which can then be reactivated.
Although conventional memory formation has not been described, TReg cells have been shown to differentiate into
terminal effector TReg cells with unique cell surface marker expression. Also contributing to the CD45RA peripheral TReg cell
compartment are converted TReg-like cells, which are derived from conventional T cells. These converted TReg-like cells
have cell surface marker expression similar to that expressed by natural TReg cells. CTLA4, cytotoxic T lymphocyte
antigen 4; FOXP3, forkhead box P3; ICOS, inducible T cell co-stimulator; IL, interleukin.

Apoptosis
A common form of cell death,
which is also known as intrinsic
or programmed cell death.
many physiological and
developmental stimuli cause
apoptosis, and this mechanism
is frequently used to delete
unwanted, superfluous or
potentially harmful cells, such
as those undergoing
transformation.

NOD/Shiscid Il2rg/ (NOG)


mice
immunodeficient mice that
can be adoptively transferred
with human cells. When
reconstituted with human cord
blood stem cells, these mice
allow the analysis of the
behaviour of human cells
in vivo.

of expression of Ki67, a nuclear proliferation marker,


indicates that these naive TReg cells are in a quiescent
stage, justifying their alternative denomination as
resting TReg cells5.
Naive TReg cells proliferate after in vitro TCR stimulation and are highly resistant to apoptosis5,, which is in
contrast to CD45RO+CD25hiFOXP3+ TReg cells, which
tend to be hyporesponsive and apoptotic after activation in vitro (see below). experiments involving transfer
of human naive TReg cells to nOD/Shi-scid Il2rg/ (nOG)
mice and in vivo TCR repertoire analysis in a healthy
donor have shown that, once activated, naive TReg cells
proliferate, upregulate FOXP3 expression and convert to CD45RO +CD25 hiFOXP3 hi TReg cells 5. These
data indicate that the human counterpart of mouse
thymus-derived natural TReg cells is more likely to be
the CD45RA+FOXP3low naive TReg cell population than
the CD45RO +CD25 hiFOXP3 + TReg cell population.
This notion is supported by the finding that almost all
FOXP3-expressing CD4+ T cells found in cord blood are
CD45RA+FOXP3low TReg cells5,66.
Although FOXP3+ recent thymic emigrants constitute a homogeneous population with a distinct phenotype (CD45RA+FOXP3lowCD31+), developing FOXP3+
thymocytes are more heterogeneous in phenotype.
Analysis of cells in the human thymus has identified

CD45RA-expressing FOXP3+ thymocytes, as well as


CD45RO- and inducible T cell co-stimulator (ICOS)expressing FOXP3+ thymocytes (discussed below)20.
CD45RO and ICOS are usually expressed by memorytype antigen-experienced T cells in the periphery and
not by recent thymic emigrants. Transient expression of
these markers might have an important role in the thymic
development of TReg cells. For instance, ICOS ligand has
been shown to be essential for the survival of ICOS+ TReg
cells in vitro20. Speculatively, the development of TReg cells
in the thymus might require transient expression of activation and/or memory markers such as CD45RO and
ICOS to avoid deletion.
Effector CD45RO+FOXP3hi TReg cells. CD45RO+FOXP3hi
T cells, which seem to derive mainly from
CD45RA+FOXP3low naive TReg cells, have potent in vitro
suppressive activity and show hyporesponsiveness following activation in vitro712. Their in vitro hyporesponsiveness, which was attributed to anergy in early studies,
is actually secondary to their high susceptibility to apoptosis following activation and during suppression5,69,70.
As shown by their expression of CD25, GITR, CD95
and CTLA4, which are all known to be upregulated by
recently activated conventional CD4+ T cells, they have
features of recently activated T cells. Importantly, similar

494 | juLy 2010 | VOLuMe 10

www.nature.com/reviews/immunol
2010 Macmillan Publishers Limited. All rights reserved

REVIEWS
Box 1 | FOXP3+ regulatory T cells throughout life
Regulatory T cells in utero
Regulatory T (TReg) cells isolated from fetal tissues express markers that are associated
with the TReg cell phenotype or with T cell activation, including CD62L, CD45RO, OX40
(also known as CD134), glucocorticoid-induced TNF-receptor-related protein (GITR),
MHC class II molecules and intracellular cytotoxic T lymphocyte antigen 4 (CTLA4)38.
These findings are of some interest given that most researchers that have
characterized TReg cells in cord blood have reported that TReg cells have a naive
phenotype, with high expression of CD45RA and low expression of CD45RO6466.
Expression of memory-type markers by fetal TReg cells may indicate activation in
peripheral tissue. This is substantiated by the presence of a small number of effector
TReg-like cells bearing a Ki67+CD45RAlowCD45RO+ FOXP3hi phenotype in cord blood5.
Recent findings indicate that fetal TReg cells have an early role in the maintenance of
fetal self tolerance and in establishing fetalmaternal tolerance99.
lifelong changes in TReg cell subsets
Analysis of naive TReg cell prevalence in peripheral blood indicates that their proportion
among CD4+ T cells declines with age, starting from a range of 410% in cord blood
and decreasing to 14% in young adults and to 0.51.5% in healthy elderly donors100.
However, the prevalence of effector TReg cells slightly increases with age starting from
a range of 00.5% in cord blood and increasing to 12.5% in young adults and to 14%
in elderly health donors5.
Because of thymic involution in adults, thymic output of T cells is greatly reduced
but still detectable in elderly people, suggesting that the involuted thymic tissue may
still be active5,66.

Thymic involution
The age-dependent decrease
of thymic epithelial volume,
which results in decreased
production of T cells.

to natural TReg cells in mice, they are highly proliferative


in vitro and in vivo, as substantiated by in vivo incorporation of tritium-labelled glucose71, Ki67 expression5 and
clonal expansion of T cells expressing particular TCRs5.
Thus, in contrast to naive TReg cells that are in a resting
state, CD45RO+FOXP3hi T cells can be thought to be an
activated and functionally differentiated subset of TReg
cells, although they must be further stimulated through
their TCR to exert suppression in vitro79,11,12. Based on
these findings, CD45RO+FOXP3hi T cells can be called
effector TReg cells. In contrast to naive TReg cells, which
are enriched in cord blood and the main subset of TReg
cells in utero38,66, effector TReg cells are more prevalent in
adults and in elderly people5 (BOX 1).
Although effector TReg cells express CD45RO, which
is a marker of conventional memory T cells, there is no
evidence for memory and recall responses by TReg cells
in mouse experiments, making it inaccurate to term
effector TReg cells a truly memory TReg cell population.
Indeed, analysis of effector TReg cell turnover in vivo
indicates that these cells undergo rapid turnover, which
is not indicative of a long-lived memory T cell population71. However, the possibility remains that effector
TReg cells may mature to become terminally differentiated suppressor cells that rapidly succumb to cell death
(see below). It remains to be determined whether the
effector TReg cell population contains a memory-type
long-lived subpopulation.
Phenotypic analysis indicates that the effector TReg
cell subset is heterogeneous in the expression of ICOS
and HLA-DR. Differential expression of ICOS has
been shown to delineate two functionally different
subsets of effector TReg cells in the periphery 20. Indeed
ICOS+ or ICOS effector TReg cells actively produce the
suppressive cytokines IL-10 or TGF, respectively 20.

Interestingly, only ICOS+ effector TReg cells could suppress the upregulation of CD86 expression on DCs, suggesting that the targets of suppression by either subset
might be different.
Similarly, the expression of HLA-DR identifies a
terminally differentiated subpopulation of effector TReg
cells. HLA-DR is expressed by approximately one third
of effector TReg cells in adult peripheral blood7. HLA-DR+
TReg cells suppress in vitro proliferation of conventional T cells and secrete cytokines more rapidly than
HLA-DR TReg cells19. Indeed, activation and expansion
of CD25hiHLA-DR TReg cells in vitro leads to the generation of HLA-DR+ TReg cells19,72. These in vitro-generated
HLA-DR+ effector TReg cells exhibit a suppressive capacity similar to that observed in ex vivo HLA-DR+ effector
TReg cells; both ex vivo-isolated and in vitro-generated
HLA-DR+ TReg cells suppress more efficiently than
HLA-DR TReg cells19. The kinetics of HLA-DR expression
by effector TReg cells is distinct from that of conventional
CD4+ T cells: conventional T cells temporarily express
HLA-DR after activation, whe reas effector TReg cells
maintain HLA-DR expression at least for thirty days post
activation19. Given these in vitro findings, it is likely that
effector TReg cells can gain and retain HLA-DR expression
in vivo. Taken together, HLA-DR+ TReg cells isolated ex vivo
seem to constitute a terminally differentiated subset in the
effector TReg cell pool (fiG. 2).

Suppressive activity of human TReg cells


In vitro suppression assay of human TReg cells. Currently,
the functional study of human TReg cell-mediated suppression is limited to in vitro co-culture assays. It should
be emphasized as a caveat that there is no direct evidence
at present that the in vitro suppression assay directly
reflects in vivo suppressive capacities of TReg cells.
Furthermore, most in vitro suppression assays measure
thymidine uptake by cultured T cells in the presence or
absence of TReg cells. This is based on the assumption
that TReg cells do not proliferate in vitro because mouse
TReg cells are consistently hyporesponsive in vitro73. This
is true for human effector CD45RAFOXP3hi TReg cells;
however, human naive CD45RA+ TReg cells actively proliferate in vitro and differentiate into CD45RA effector
TReg cells while suppressing effector cell proliferation.
This indicates that thymidine uptake by cultured TReg
and effector T cells is not appropriate to assess suppressive activity of human naive CD45RA+ TReg cells.
we propose that it is more accurate to assess dilution
of 5,6-carboxyfluorescein diacetate succinimidyl ester
(CFSe) in effector T cells to determine the percentage
and number of proliferating cells5.
The precise molecular mechanisms of suppression
by human TReg cells remains to be determined, although
in vitro and in vivo mouse studies have implicated several mechanisms (TABle 2). These include modulation of
the cytokine microenvironment, metabolic disruption
of the target cell, alteration of DC activating capacity and
cytolysis (reviewed in RefS 1,74,75). For example, human
TReg cells can be suppressive in vitro even in the absence
of APCs7, indicating that target cell suppression can occur
through direct contact between TReg and effector T cells.

NATuRe ReVIewS | Immunology

VOLuMe 10 | juLy 2010 | 495


2010 Macmillan Publishers Limited. All rights reserved

REVIEWS
Table 2 | TReg cell suppressive mechanisms
Key molecule(s)

Function

Refs
mouse

Human

Mechanisms of contact-dependent suppression


CTLA4

Downregulation of APC co-stimulatory function

76

Interaction with CD80 and CD86 on conventional T cells

101

CD73CD39

Hydrolysis of inflammatory extracellular ATP

102

LAG3

Induction of inhibitory signalling through MHC class II molecules

103

Granzyme B (mouse) and


granzyme A (human)

Lysis of conventional T cells

104,105

106

CD95CD95 ligand

Induction of apoptosis in conventional T cells

107

108,109

Mechanisms of cytokine-mediated suppression


TGF and LAP

Induction of FOXP3 in conventional T cells

IL-10

Attentuation of DC function
Conversion of conventional T cells to TR1 cells

20

110,111

111

Galectin 1

Cell cycle arrest and apoptosis in conventional T cells

112

CD25

Adsorption of IL-2

113

IL-35

Induction of conventional T cell expression of IL-35 by TReg cells


enhances suppression (IL-35 is not expressed by human TReg cells)

114

115

APC, antigen-presenting cell; CTLA4, cytotoxic T lymphocyte antigen 4; DC, dendritic cell; FOXP3, forkhead box P3; IL, interleukin;
LAG3, lymphocyte activation gene 3; LAP, latency-associated peptide; TGF, transforming growth factor-; TReg, regulatory T.

A recent study in mice has shown that CTLA4 is crucial


for the suppressive function of FOXP3+ TReg cells both
in vitro and in vivo76. CTLA4 expressed by TReg cells can
modulate CD80 and CD86 expression by DCs and thereby
inhibit the activation of effector T cells. In humans, it is
intriguing that only CD45RO+FOXP3hi effector TReg cells
express CTLA4 at high levels5. It remains to be determined
whether the action of CTLA4 on APCs is indispensable
for the in vitro suppressive function of TReg cells.
TCR signal strength and cytokine milieu. with the caveat
regarding in vitro suppression assay discussed above,
culture conditions that allow human TReg cells to exert
in vitro suppressive activity have been characterized.
Human TReg cells must be activated through their
TCR to be functionally suppressive79,11,12. Once activated,
they seemingly do not need to be viable in co-culture
in order to mediate suppression, as TReg cells fixed with
paraformaldehyde following activation remain suppressive77. This correlates with the finding that most effector
TReg cells are apoptotic following in vitro TCR stimulation but are still highly suppressive5. These data suggest
that human TReg cells undergo activation-induced modulation of specific cell surface molecules and that this
change, without further alteration of TReg cell function,
can mediate suppression.
The effectiveness of suppression also depends on the
quality of T cell stimulation, as the strength of the effector T cell stimulation has a strong influence on whether
suppression ensues. effector T cells activated in the
presence of strong co-stimulatory signals are refractory
to TReg cell-mediated suppression, as are effector T cells
supplemented with growth-promoting cytokines7,77.
Increasing the strength of the TCR signal received by

effector T cells in culture can also increase their resistance to suppression7. These findings suggest that human
TReg cells probably cannot suppress pro-inflammatory
cytokine production and proliferation in conditions in
which effector T cells are strongly activated.
In addition, in pro-inflammatory microenvironments, human TReg cells can be induced to secrete the
pro-inflammatory cytokine IL-17 (Ref. 78). IL-17+FOXP3+
TReg cells have been isolated from human peripheral
blood5,78,79 and are suppressive in co-culture assays in
the presence of low TCR stimulation, but they concomitantly lose the ability to suppress and gain the capacity to secrete IL-17 when they are strongly activated in
the presence of the pro-inflammatory cytokines IL-1
and IL-6 (Ref. 78). It is likely that the loss of suppression
observed in the presence of strongly activating factors is
due to both an increase in effector T cells resistance and
a decrease in TReg cell function.

Clinical applications for TReg cell biology


Dysfunctional TReg cells can be a cause of autoimmune
disease, allergy and immunopathology. But it remains to
be determined how these cells are involved in the pathophysiology of common immunological diseases such as
autoimmune disease2,80. They have a role in the maintenance of allograft and fetalmaternal tolerance and the
control of various immune responses; for example, to
cancer cells and microorganisms1. However, it has yet
to be established how to reliably monitor the contribution of TReg cells to tolerance and immune control. with
recent data that FOXP3+ T cells are heterogeneous and
include regulatory and non-regulatory T cell subsets, it
is important and necessary to quantitatively and qualitatively assess each subpopulation of FOXP3+ T cells, rather

496 | juLy 2010 | VOLuMe 10

www.nature.com/reviews/immunol
2010 Macmillan Publishers Limited. All rights reserved

REVIEWS
than enumeration of the whole FOXP3+ T cell population
and in vitro functional examination of CD25hi TReg cells.
Indeed there are disease-specific patterns in the composition of naive and effector FOXP3+ TReg cells and nonregulatory FOXP3 + T cells 5. Targeting a particular
FOXP3+ TReg cell subpopulation, rather than all FOXP3+
T cells, makes the control of immune responses more
effective: for example, in cell therapy with TReg cells, their
in vivo expansion to suppress immune responses and
TReg cell depletion or functional blockade to enhance
immune responses.

Rapamycin
An immunosuppressive drug
that does not prevent T cell
activation but blocks
il-2-mediated clonal
expansion by blocking mTOR
(mammalian target of
rapamycin).

Deacetylation
A post-translational
modification of chromatin
components, particularly
histones. it correlates with
actively transcribed chromatin.
Histone deacetylases have
been identified as components
of nuclear co-repressor
complexes, which reverse the
actions of histone
acetyltransferases, thereby
inhibiting gene transcription.

Immunosuppression by TReg cell therapy. Cellular therapy


based on ex vivo expansion of TReg cells and their transfer
to patients is currently the focus of intense research to
treat autoimmune disease and inhibit the occurrence of
graft-versus-host disease after bone marrow transplantation81,82. Transfer of expanded TReg cells is effective in the
prevention and, in some cases, the treatment of ongoing inflammatory and autoimmune diseases in mice1.
However, several key issues need to be addressed before
their clinical use.
First, because of the heterogeneity of FOXP3 +
T cells, the TReg cell population that should be isolated
for expansion must be determined. The expansion
of FOXP3-expressing T cells, isolated based on the
expression of CD25 and CD127, needs to be undertaken with caution because this population contains
non-regulatory CD45RO+FOXP3low T cells that produce pro-inflammatory cytokines and may constitute
3050% of the FOXP3+CD4+ T cells5,72. Furthermore,
several reports have indicated that CD45RA+FOXP3low
naive TReg cells have the highest capacity to maintain
FOXP3 expression following expansion, making this
subset the population of choice for isolation5,83.
Second, it has been shown that effector TReg cells are
prone to die by apoptosis5,69 and expand poorly even
in cultures with high-dose IL-2 (Ref. 72). By contrast,
naive TReg cells can be easily expanded and develop a
high frequency of functional FOXP3+ TReg cells, which
again supports the use of CD45RA+FOXP3low naive TReg
cells as the subset of choice for TReg cell expansion84.
Nevertheless, even adequately purified naive TReg cells,
when cultured in a facilitating cytokine milieu containing high-dose IL-2, can give rise to a substantial fraction
of T cells that produce pro-inflammatory cytokines84.
Thus, it is important to develop and establish a cocktail
of cytokines and chemicals that may enable the expansion of 100% pure functional TReg cells that do not secrete
pro-inflammatory cytokines. Rapamycin (Sirolimus/
Rapamune; wyeth), which substantially increases TReg
cell purity by eliminating non-TReg cells85, may be useful
as a component of this cocktail.
Third, even if 100% purity is achieved in vitro,
in vivo stability of infused TReg cells, especially under
pro-inflammatory conditions, is not guaranteed. Indeed,
in mice, recent reports have highlighted the plasticity of
TReg cells towards effector TH1, TH2 or TH17 cell phenotypes under pathological conditions86,87. Although data
in mice indicate that transferred TReg cells can survive for
several months67, it remains to be determined how long

infused TReg cells can survive in humans and whether


a single infusion of expanded TReg cells is sufficient to
have therapeutic effects. In addition, as stable FOXP3
expression is a prerequisite for the suppressive function
of TReg cells, functional stability of infused FOXP3+ TReg
cells could be maintained or even enhanced by FOXP3
gene histone deacetylation or promotion of the acetylation of FOXP3 protein itself by inhibitors of histone
deacetylase88 such as vorinostat (also known as suberoylanilide hydroxamic acid (Zolinza; Merck)). The drug is
in clinical use for the management of cutaneous T cell
lymphoma but it remains to be determined whether it
can have a TReg cell-specific effect in vivo89.
Thus, successful cell therapy using FOXP3+ TReg cells
depends on how TReg cells are prepared as a pure population and how they can be maintained as a functionally
stable population in vivo and also in vitro when they are
expanded in an antigen-specific manner. Further study
is also required regarding how functionally competent
antigen-specific TReg cells can be induced from naive
T cells in vitro51.
Immune potentiation by targeting TReg cells. There is
accumulating evidence that FOXP3+ TReg cells impede
the development of effective tumour immunity in cancerbearing hosts and antimicrobial immunity in individuals
with chronic infection.
It has been documented that a large number of
CD25+CD4+ TReg cells are present in tumours and draining lymph nodes in patients with cancer. In particular,
decreased ratios of CD8+ T cells to FOXP3+CD25+CD4+
TReg cells in tumours correlate with poor prognosis90. It
has also been shown in numerous mouse models that
depletion of TReg cells enhances antitumour immune
responses, leading to the eradication of tumours91.
Similarly, in humans, recent studies have shown that
tumour antigen-specific CD4+ T cells can expand in
patients with cancer and healthy individuals following in vitro antigenic stimulation of peripheral CD4+
T cells isolated from the individual after depletion of
CD25+CD4+ T cells121.
Based on these findings, efforts have been made to find
cell surface molecules that are predominantly expressed
by TReg cells or can specifically modulate TReg cell function.
use of monoclonal antibodies specific for such molecules
have revealed that monoclonal antibodies specific for
CD25 (a depleting antibody), CTLA4 (a blocking antibody), GITR (an agonistic antibody) and OX40 (an agonistic antibody) can enhance tumour immunity in mice.
Small molecules, such as cyclophosphamide, that deplete
TReg cells can also enhance tumour-specific immunity122.
CTLA4-specific blocking monoclonal antibodies (such as
ipilimumab (MDX-010; Bristol-Myers Squibb/Medarex),
are now in clinical use to treat advanced stages of cancer 92. Since many TReg cell-specific cell surface markers are
also expressed by activated T cells, combinations of these
monoclonal antibodies that target different molecules
may be more effective in controlling the balance between
TReg cells and effector T cells towards dominance of the
effector T cells.
The contribution of TReg cells to infectious diseases

NATuRe ReVIewS | Immunology

VOLuMe 10 | juLy 2010 | 497


2010 Macmillan Publishers Limited. All rights reserved

REVIEWS
Table 3 | Clinical application of TReg cells
Application

In use

under investigation

Side effects and


caveats

Infection

Molecules inhibiting TReg cell function,


such as CTLA4-specific antibody
(ipilimumab (MDX-010; Bristol-Myers
Squibb/Medarex))

Molecules inhibiting TReg cell


function and differentiation

Low efficacy
in infectious
diseases

Cancer

Molecules that deplete TReg cell


populations, such as DAB389IL-2
(denileukin difitox (Ontak; Eisai))

Molecules that deplete TReg cells

Induction of
autoimmunity

Autoimmunity

Molecules mimicking TReg cell-mediated


suppression, such as CTLA4Ig (Abatacept
(Orencia; Bristol-Myers Squibb))

Cellular therapy based on


TReg cell expansion*

Suppression
of cancer
surveillance and
immunity

Molecules that favour TReg cell survival


and development, such as rapamycin
(Sirolimus/Rapamune; Wyeth)

Molecules capable of
maintaining or improving
TReg cell purity and survival

Allergy
Pregnancy
Transplantation*

Molecules mimicking TReg


cell-mediated suppression

Feasibility during
pregnancy not
evaluated

CTLA4, cytotoxic T lymphocyte antigen 4; Ig, immunoglobulin; TReg, regulatory T. *In limited use for some patients with
graft-versus-host disease82.

is rather complex. Depletion of TReg cells can enhance


an antimicrobial response and eradicate microorganisms. By contrast, TReg cells suppress antimicrobial
T cell responses to a certain degree, allowing a certain
amount of microorganisms to survive and thereby allow
memory T cells to develop93. They are also directly
involved in anti-infectious responses as facilitators
for the early entry of effector T cells into infected tissues94. In humans, TReg cell numbers have been shown
to increase in early studies of chronic viral infections
such as chronic hepatitis C95 and HIV infections96 and
also in severe conditions such as septic shock97. Similar
to immunotherapy of cancer, antimicrobial immune
responses can be controlled by differential targeting of
TReg cells and effector T cells.
Together, TReg cell-based therapies are a promising
strategy for the management of various immunological
diseases and immune responses (TABle 3).

Box 2 | Issues to address for human TReg cells


What are the respective roles of naive and effector regulatory T (TReg) cells in the
maintenance of self tolerance in humans?
What is the relationship between effector TReg cell subsets (HLA-DR+ and HLA-DR
TReg cells and inducible T cell co-stimulator (ICOS)+ and ICOS TReg cells)?
Do induced TReg cells have a specific phenotype compared with effector TReg cells?
Do in vitro suppression assays directly reflect the suppressive capacity of human
TReg cells in vivo?
What is the main molecular mechanism (or mechanisms) of TReg cell-mediated
suppression in humans?
How stable are TReg cell subsets throughout life? Do TReg cells make memory and
recall responses? How variable is the T cell receptor repertoire of TReg subsets
throughout life?
Can we determine a combination of molecules that can maintain TReg cell stability
and purity in vitro and in vivo, thus preventing conversion of TReg cells into
pathogenic T cells?
The discovery of a highly specific TReg cell surface marker is required to provide a
highly targeted depleting monoclonal antibody for the management of cancer.

Conclusion
Since the description of suppressor T cells in the early
1970s, considerable progress has been made in the
characterization of TReg cells. It is now well established
that human FOXP3+CD4+ T cells can be divided into
subpopulations based on their expression of cell surface
markers such as CD45RA and CD45RO and the level
of FOXP3 expression. Following antigenic stimulation,
CD45RA+FOXP3low naive TReg cells seem to differentiate into CD45RO+FOXP3hi effector TReg cells, which
exert strong suppression and then die by apoptosis. The
FOXP3+ T cell population also include non-regulatory
T cells, precluding the use of FOXP3 as a sole marker for
human TReg cells.
It remains to be determined how TReg cells, in particular effector TReg cells, exert suppression in vitro and
in vivo. TReg cells may use different mechanisms of suppression concurrently and sequentially. However, it is
still possible that a core mechanism of suppression that
is commonly used by every TReg cell at any location or
in any environment exists75,98. In addition, given the
memory-like phenotype of effector TReg cells, it is not
clear whether TReg cells mount a memory response and,
if it is the case, where the resting niche for these resident
memory TReg cells would be.
For TReg cell-based therapy in humans, the key issue
to address remains the stability of TReg cells in vitro and
in vivo. Because of their possible plasticity towards
pathogenic T H1, T H2 or T H17 cell phenotypes, it is
mandatory to determine the correct combination of
molecules that would maintain the purity and the stability of TReg cells in vitro during and/or after expansion
and after their transfer into patients. Identification of
a highly specific TReg cell surface marker is still needed
for tightly targeted depletion of TReg cells for the treatment of cancer. Although several issues regarding longterm reliability and safety need to be addressed, TReg
cell-based therapy is a promising therapeutic modality that is applicable to a wide range of immunological
diseases (BOX 2).

498 | juLy 2010 | VOLuMe 10

www.nature.com/reviews/immunol
2010 Macmillan Publishers Limited. All rights reserved

REVIEWS
1.
2.
3.
4.
5.

6.
7.

8.
9.

10.
11.

12.

13.

14.
15.

16.
17.

18.

19.

20.

21.

Sakaguchi, S., Yamaguchi, T., Nomura, T. & Ono, M.


Regulatory T cells and immune tolerance. Cell 133,
775787 (2008).
BaecherAllan, C. & Hafler, D. A. Human regulatory
T cells and their role in autoimmune disease.
Immunol. Rev. 212, 203216 (2006).
Sakaguchi, S., Wing, K. & Miyara, M. Regulatory
T cells a brief history and perspective.
Eur. J. Immunol. 37, S116S123 (2007).
Costantino, C., BaecherAllan, C. & Hafler, D.
Human regulatory T cells and autoimmunity.
Eur. J. Immunol. 38, 921924 (2008).
Miyara, M. et al. Functional delineation and
differentiation dynamics of human CD4+ T cells
expressing the FoxP3 transcription factor.
Immunity 30, 899911 (2009).
This paper provides a phenotypic and functional
characterization of human FOXP3+ TReg cell subsets
and shows that naive TReg cells differentiate into
effector TReg cells.
Jiang, H. & Chess, L. Regulation of immune responses
by T cells. N. Engl. J. Med. 354, 11661176 (2006).
BaecherAllan, C., Brown, J. A., Freeman, G. J. &
Hafler, D. A. CD4+CD25high regulatory cells in human
peripheral blood. J. Immunol. 167, 12451253
(2001).
Taams, L. S. et al. Antigenspecific T cell suppression
by human CD4+CD25+ regulatory T cells.
Eur. J. Immunol. 32, 16211630 (2002).
Levings, M. K., Sangregorio, R. & Roncarolo, M. G.
Human CD25+CD4+ T regulatory cells suppress naive
and memory T cell proliferation and can be expanded
in vitro without loss of function. J. Exp. Med. 193,
12951302 (2001).
Ng, W. F. et al. Human CD4+CD25+ cells: a naturally
occurring population of regulatory T cells. Blood 98,
27362744 (2001).
Jonuleit, H. et al. Identification and functional
characterization of human CD4+CD25+ T cells with
regulatory properties isolated from peripheral blood.
J. Exp. Med. 193, 12851294 (2001).
Dieckmann, D., Plottner, H., Berchtold, S., Berger, T.
& Schuler, G. Ex vivo isolation and characterization of
CD4+CD25+ T cells with regulatory properties from
human blood. J. Exp. Med. 193, 13031310 (2001).
References 712 show that, similar to mouse
CD25+CD4+ T cells, human CD25+CD4+ T cells
have suppressive properties in vitro. Of note,
reference 7 shows that CD4+ T cells with the
highest expression of CD25 have the strongest
suppressive activity, that is, contain pure TReg cells.
Sakaguchi, S., Sakaguchi, N., Asano, M., Itoh, M. &
Toda, M. Immunologic selftolerance maintained by
activated T cells expressing IL2 receptor chains
(CD25). Breakdown of a single mechanism of self
tolerance causes various autoimmune diseases.
J. Immunol. 155, 11511164 (1995).
Khattri, R., Cox, T., Yasayko, S. A. & Ramsdell, F.
An essential role for Scurfin in CD4+CD25+ T
regulatory cells. Nature Immunol. 4, 337342 (2003).
Fontenot, J. D., Gavin, M. A. & Rudensky, A. Y. Foxp3
programs the development and function of
CD4+CD25+ regulatory T cells. Nature Immunol.
4, 330336 (2003).
Hori, S., Nomura, T. & Sakaguchi, S. Control of
regulatory T cell development by the transcription
factor Foxp3. Science 299, 10571061 (2003).
Roncador, G. et al. Analysis of FOXP3 protein
expression in human CD4+CD25+ regulatory T cells at
the singlecell level. Eur. J. Immunol. 35, 16811691
(2005).
Gavin, M. A. et al. Singlecell analysis of normal and
FOXP3mutant human T cells: FOXP3 expression
without regulatory T cell development. Proc. Natl
Acad. Sci. USA 103, 66596664 (2006).
This manuscript first reported that naive CD4+
T cells can upregulate FOXP3 expression at the
protein level following in vitro activation.
BaecherAllan, C., Wolf, E. & Hafler, D. MHC class II
expression identifies functionally distinct human
regulatory T cells. J. Immunol. 176, 46224631
(2006).
Ito, T. et al. Two functional subsets of FOXP3+
regulatory T cells in human thymus and periphery.
Immunity 28, 870880 (2008).
References 19 and 20 provide further information
on the phenotype of effector TReg cells, that is, the
expression of HLA-DR and ICOS.
Itoh, M. et al. Thymus and autoimmunity: production
of CD25+CD4+ naturally anergic and suppressive
T cells as a key function of the thymus in maintaining

22.

23.
24.

25.

26.

27.

28.
29.
30.
31.
32.
33.

34.

35.
36.

37.
38.

39.

40.

41.

42.

43.
44.
45.

46.

immunologic selftolerance. J. Immunol. 162,


53175326 (1999).
Fontenot, J. D., Dooley, J. L., Farr, A. G. &
Rudensky, A. Y. Developmental regulation of Foxp3
expression during ontogeny. J. Exp. Med. 202,
901906 (2005).
Lahl, K. et al. Selective depletion of Foxp3+ regulatory
T cells induces a scurfylike disease. J. Exp. Med. 204,
5763 (2007).
Fontenot, J. D., Gavin, M. A. & Rudensky, A. Y.
Foxp3 programs the development and function of
CD4+CD25+ regulatory T cells. Nature Immunol.
4, 330336 (2003).
Fattorossi, A. et al. Thymopoiesis, regulatory T cells,
and TCRV expression in thymoma with and without
myasthenia gravis, and modulatory effects of steroid
therapy. J. Clin. Immunol. 28, 194206 (2008).
Tuovinen, H., Kekalainen, E., Rossi, L. H., Puntila, J. &
Arstila, T. P. Cutting edge: human CD4CD8
thymocytes express FOXP3 in the absence of a TCR.
J. Immunol. 180, 36513654 (2008).
Tuovinen, H., Pekkarinen, P. T., Rossi, L. H., Mattila, I.
& Arstila, T. P. The FOXP3+ subset of human
CD4+CD8+ thymocytes is immature and subject to
intrathymic selection. Immunol. Cell Biol. 86,
523529 (2008).
Lee, H. M. & Hsieh, C. S. Rare development of Foxp3+
thymocytes in the CD4+CD8+ subset. J. Immunol.
183, 22612266 (2009).
Liston, A. et al. Differentiation of regulatory Foxp3+
T cells in the thymic cortex. Proc. Natl Acad. Sci. USA
105, 1190311908 (2008).
Picca, C. C. et al. Role of TCR specificity in CD4+
CD25+ regulatory Tcell selection. Immunol. Rev. 212,
7485 (2006).
Malek, T. R. et al. IL2 family of cytokines in T
regulatory cell development and homeostasis.
J. Clin. Immunol. 28, 635639 (2008).
Spits, H. Development of T cells in the human
thymus. Nature Rev. Immunol. 2, 760772 (2002).
Watanabe, N. et al. Human thymic stromal
lymphopoietin promotes dendritic cellmediated CD4+
T cell homeostatic expansion. Nature Immunol.
5, 426434 (2004).
Hanabuchi, S. et al. Thymic stromal lymphopoietin
activated plasmacytoid dendritic cells induce the
generation of FOXP3+ regulatory T cells in human
thymus. J. Immunol. 184, 29993007 (2010).
Watanabe, N. et al. Hassalls corpuscles instruct
dendritic cells to induce CD4+CD25+ regulatory T cells
in human thymus. Nature 436, 11811185 (2005).
Liu, Y. J. et al. TSLP: an epithelial cell cytokine that
regulates T cell differentiation by conditioning
dendritic cell maturation. Annu. Rev. Immunol. 25,
193219 (2007).
Stites, D. P. & Pavia, C. S. Ontogeny of human T cells.
Pediatrics 64, 795802 (1979).
DarrasseJeze, G., Marodon, G., Salomon, B. L.,
Catala, M. & Klatzmann, D. Ontogeny of CD4+CD25+
regulatory/suppressor T cells in human fetuses.
Blood 105, 47154721 (2005).
Brunkow, M. E. et al. Disruption of a new forkhead/
wingedhelix protein, scurfin, results in the fatal
lymphoproliferative disorder of the scurfy mouse.
Nature Genet. 27, 6873 (2001).
Wildin, R. S. et al. Xlinked neonatal diabetes mellitus,
enteropathy and endocrinopathy syndrome is the
human equivalent of mouse scurfy. Nature Genet. 27,
1820 (2001).
Bennett, C. L. et al. The immune dysregulation,
polyendocrinopathy, enteropathy, Xlinked syndrome
(IPEX) is caused by mutations of FOXP3. Nature
Genet. 27, 2021 (2001).
Yagi, H. et al. Crucial role of FOXP3 in the
development and function of human CD25+CD4+
regulatory T cells. Int. Immunol. 16, 16431656
(2004).
Buckner, J. H. & Ziegler, S. F. Functional analysis of
FOXP3. Ann. N. Y Acad. Sci. 1143, 151169 (2008).
Allan, S. E. et al. The role of 2 FOXP3 isoforms in the
generation of human CD4+ Tregs. J. Clin. Invest. 115,
32763284 (2005).
AartsRiemens, T., Emmelot, M. E., Verdonck, L. F. &
Mutis, T. Forced overexpression of either of the two
common human Foxp3 isoforms can induce regulatory
T cells from CD4+CD25 cells. Eur. J. Immunol. 38,
13811390 (2008).
Du, J., Huang, C., Zhou, B. & Ziegler, S. F. Isoform
specific inhibition of RORmediated transcriptional
activation by human FOXP3. J. Immunol. 180,
47854792 (2008).

NATuRe ReVIewS | Immunology

47. Bettelli, E., Dastrange, M. & Oukka, M. Foxp3


interacts with nuclear factor of activated T cells and
NFB to repress cytokine gene expression and
effector functions of T helper cells. Proc. Natl Acad.
Sci. USA 102, 51385143 (2005).
48. Lopes, J. E. et al. Analysis of FOXP3 reveals multiple
domains required for its function as a transcriptional
repressor. J. Immunol. 177, 31333142 (2006).
49. Walker, M. R. et al. Induction of FoxP3 and
acquisition of T regulatory activity by stimulated
human CD4+CD25 T cells. J. Clin. Invest. 112,
14371443 (2003).
This paper reports that naive CD4+ T cells can
upregulate FOXP3 expression at the mRNA level
in vitro following activation.
50. Walker, M. R., Carson, B. D., Nepom, G. T., Ziegler, S. F.
& Buckner, J. H. De novo generation of antigen
specific CD4+CD25+ regulatory T cells from human
CD4+CD25 cells. Proc. Natl Acad. Sci. USA 102,
41034108 (2005).
51. Allan, S. E. et al. Generation of potent and stable
human CD4+ T regulatory cells by activation
independent expression of FOXP3. Mol. Ther. 16,
194202 (2008).
52. Hoffmann, P. et al. Loss of FOXP3 expression in
natural human CD4+CD25+ regulatory T cells upon
repetitive in vitro stimulation. Eur. J. Immunol. 39,
10881097 (2009).
53. Allan, S. E., SongZhao, G. X., Abraham, T.,
McMurchy, A. N. & Levings, M. K. Inducible
reprogramming of human T cells into Treg cells by a
conditionally active form of FOXP3. Eur. J. Immunol.
38, 32823289 (2008).
54. Coombes, J. L. et al. A functionally specialized
population of mucosal CD103+ DCs induces Foxp3+
regulatory T cells via a TGF and retinoic acid
dependent mechanism. J. Exp. Med. 204,
17571764 (2007).
55. Hill, J. A. et al. Foxp3 transcriptionfactordependent
and independent regulation of the regulatory T cell
transcriptional signature. Immunity 27, 786800
(2007).
56. Feuerer, M., Hill, J. A., Mathis, D. & Benoist, C.
Foxp3+ regulatory T cells: differentiation,
specification, subphenotypes. Nature Immunol. 10,
689695 (2009).
57. Tran, D. Q., Ramsey, H. & Shevach, E. M. Induction of
FOXP3 expression in naive human CD4+FOXP3 T cells
by Tcell receptor stimulation is transforming growth
factor dependent but does not confer a regulatory
phenotype. Blood 110, 29832990 (2007).
58. Allan, S. E. et al. Activationinduced FOXP3 in human
T effector cells does not suppress proliferation or
cytokine production. Int. Immunol. 19, 345354
(2007).
59. Liu, W. CD127 expression inversely correlates with
FoxP3 and suppressive function of human CD4+ T reg
cells. J. Exp. Med. 203, 17011711 (2006).
60. Seddiki, N. et al. Expression of interleukin (IL)2 and
IL7 receptors discriminates between human
regulatory and activated T cells. J. Exp. Med. 203,
1693 (2006).
References 59 and 60 show that separation of
CD4+ T cells based on expression of low levels of
CD127 combined with high levels of CD25 enables
the isolation of FOXP3-expressing CD4+ T cells.
61. Mazzucchelli, R. & Durum, S. K. Interleukin7 receptor
expression: intelligent design. Nature Rev. Immunol.
7, 144154 (2007).
62. Aerts, N. E. et al. Activated T cells complicate the
identification of regulatory T cells in rheumatoid
arthritis. Cell. Immunol. 251, 109115 (2008).
63. Hamann, A., Klugewitz, K., Austrup, F. &
JablonskiWestrich, D. Activation induces rapid and
profound alterations in the trafficking of T cells.
Eur. J. Immunol. 30, 32073218 (2000).
64. Seddiki, N. et al. Persistence of naive CD45RA+
regulatory T cells in adult life. Blood 107, 28302838
(2006).
65. Valmori, D., Merlo, A., Souleimanian, N. E.,
Hesdorffer, C. S. & Ayyoub, M. A peripheral
circulating compartment of natural naive CD4 Tregs.
J. Clin. Invest. 115, 19531962 (2005).
66. Fritzsching, B. et al. Naive regulatory T cells: a novel
subpopulation defined by resistance toward CD95L
mediated cell death. Blood 108, 33713378 (2006).
References 64, 65 and 66 describe the phenotype
and the function of naive TReg cells.
67. Fisson, S. et al. Continuous activation of autoreactive
CD4+ CD25+ regulatory T cells in the steady state.
J. Exp. Med. 198, 737746 (2003).

VOLuMe 10 | juLy 2010 | 499


2010 Macmillan Publishers Limited. All rights reserved

REVIEWS
68. Brod, S. A., Rudd, C. E., Purvee, M. & Hafler, D. A.
Lymphokine regulation of CD45R expression on human
T cell clones. J. Exp. Med. 170, 21472152 (1989).
69. Fritzsching, B. et al. In contrast to effector T cells,
CD4+CD25+FoxP3+ regulatory T cells are highly
susceptible to CD95 ligand but not to TCRmediated
cell death. J. Immunol. 175, 3236 (2005).
70. Ashley, C. W. & BaecherAllan, C. Cutting Edge:
Responder T cells regulate human DR+ effector
regulatory T cell activity via granzyme B. J. Immunol.
183, 48434847 (2009).
71. VukmanovicStejic, M. et al. Human CD4+ CD25hi
Foxp3+ regulatory T cells are derived by rapid
turnover of memory populations in vivo. J. Clin. Invest.
116, 24232433 (2006).
72. Putnam, A. et al. Expansion of human regulatory
Tcells from patients with type 1 diabetes. Diabetes
58, 652662 (2008).
73. Thornton, A. M. & Shevach, E. M. CD4+CD25+
immunoregulatory T cells suppress polyclonal T cell
activation in vitro by inhibiting interleukin 2
production. J. Exp. Med. 188, 287296 (1998).
74. Vignali, D. A., Collison, L. W. & Workman, C. J.
How regulatory T cells work. Nature Rev. Immunol.
8, 523532 (2008).
75. Shevach, E. M. Mechanisms of foxp3+ T regulatory
cellmediated suppression. Immunity 30, 636645
(2009).
76. Wing, K. et al. CTLA4 control over Foxp3+ regulatory
T cell function. Science 322, 271275 (2008).
77. BaecherAllan, C., Viglietta, V. & Hafler, D. A.
Inhibition of human CD4+CD25+high regulatory T cell
function. J. Immunol. 169, 62106217 (2002).
78. Beriou, G. et al. IL17 producing human peripheral
regulatory T cells retain suppressive function. Blood
30, 42404249 (2009).
79. Ayyoub, M. et al. Human memory FOXP3+ Tregs
secrete IL17 ex vivo and constitutively express the
TH17 lineagespecific transcription factor RORt.
Proc. Natl Acad. Sci. USA 106, 86358640 (2009).
80. Bacchetta, R., Gambineri, E. & Roncarolo, M. G.
Role of regulatory T cells and FOXP3 in human
diseases. J. Allergy Clin. Immunol. 120, 227235
(2007).
81. Miyara, M., Wing, K. & Sakaguchi, S. Therapeutic
approaches to allergy and autoimmunity based on
FoxP3+ regulatory Tcell activation and expansion.
J. Allergy Clin. Immunol. 123, 749755 (2009).
82. Trzonkowski, P. et al. Firstinman clinical results of the
treatment of patients with graft versus host disease
with human ex vivo expanded CD4+CD25+CD127
T regulatory cells. Clin. Immunol. 133, 2226 (2009).
83. Hoffmann, P., Eder, R., KunzSchughart, L. A.,
Andreesen, R. & Edinger, M. Largescale in vitro
expansion of polyclonal human CD4+CD25high
regulatory T cells. Blood 104, 895903 (2004).
84. Hoffmann, P. et al. Only the CD45RA+ subpopulation
of CD4+CD25high T cells gives rise to homogeneous
regulatory Tcell lines upon in vitro expansion. Blood
108, 42604267 (2006).
85. Battaglia, M. et al. Rapamycin promotes expansion of
functional CD4+CD25+FOXP3+ regulatory T cells of
both healthy subjects and type 1 diabetic patients.
J. Immunol. 177, 83388347 (2006).
86. Wan, Y. Y. & Flavell, R. A. Regulatory Tcell functions
are subverted and converted owing to attenuated
Foxp3 expression. Nature 445, 766770 (2007).
87. Zhou, X. et al. Instability of the transcription factor
Foxp3 leads to the generation of pathogenic memory
T cells in vivo. Nature Immunol. 10, 10001007 (2009).
88. Tao, R. et al. Deacetylase inhibition promotes the
generation and function of regulatory T cells.
Nature Med. 13, 12991307 (2007).
89. Mann, B. S., Johnson, J. R., Cohen, M. H., Justice, R.
& Pazdur, R. FDA approval summary: vorinostat for
treatment of advanced primary cutaneous Tcell
lymphoma. Oncologist 12, 12471252 (2007).

90. Curiel, T. J. et al. Specific recruitment of regulatory


T cells in ovarian carcinoma fosters immune privilege
and predicts reduced survival. Nature Med. 10,
942949 (2004).
91. Colombo, M. P. & Piconese, S. RegulatoryTcell
inhibition versus depletion: the right choice in cancer
immunotherapy. Nature Rev. Cancer 7, 880887
(2007).
92. Hodi, F. S. et al. Immunologic and clinical effects of
antibody blockade of cytotoxic T lymphocyte
associated antigen 4 in previously vaccinated cancer
patients. Proc. Natl Acad. Sci. USA 105, 30053010
(2008).
93. Belkaid, Y., Piccirillo, C. A., Mendez, S., Shevach, E. M.
& Sacks, D. L. CD4+CD25+ regulatory T cells control
Leishmania major persistence and immunity. Nature
420, 502507 (2002).
94. Lund, J. M., Hsing, L., Pham, T. T. & Rudensky, A. Y.
Coordination of early protective immunity to viral
infection by regulatory T cells. Science 320,
12201224 (2008).
95. Boettler, T. et al. T cells with a CD4+CD25+
regulatory phenotype suppress in vitro proliferation
of virusspecific CD8+ T cells during chronic hepatitis
C virus infection. J. Virol. 79, 78607867 (2005).
96. Weiss, L. et al. Human immunodeficiency virus
driven expansion of CD4+CD25+ regulatory T cells,
which suppress HIVspecific CD4 Tcell responses in
HIVinfected patients. Blood 104, 32493256
(2004).
97. Venet, F. et al. Increased percentage of CD4+CD25+
regulatory T cells during septic shock is due to the
decrease of CD4+CD25 lymphocytes. Crit. Care Med.
32, 23292331 (2004).
98. Sakaguchi, S., Wing, K., Onishi, Y., PrietoMartin, P.
& Yamaguchi, T. Regulatory T cells: how do they
suppress immune responses? Int. Immunol. 21,
11051111 (2009).
99. Mold, J. E. et al. Maternal alloantigens promote the
development of tolerogenic fetal regulatory T cells
in utero. Science 322, 15621565 (2008).
100. Takahata, Y. et al. CD25+CD4+ T cells in human cord
blood: an immunoregulatory subset with naive
phenotype and specific expression of forkhead box
p3 (Foxp3) gene. Exp. Hematol. 32, 622629
(2004).
101. Paust, S., Lu, L., McCarty, N. & Cantor, H.
Engagement of B7 on effector T cells by regulatory
T cells prevents autoimmune disease. Proc. Natl Acad.
Sci. USA 101, 1039810403 (2004).
102. Deaglio, S. et al. Adenosine generation catalyzed by
CD39 and CD73 expressed on regulatory T cells
mediates immune suppression. J. Exp. Med. 204,
12571265 (2007).
103. Huang, C. T. et al. Role of LAG3 in regulatory T cells.
Immunity 21, 503513 (2004).
104. Gondek, D. C., Lu, L. F., Quezada, S. A., Sakaguchi, S.
& Noelle, R. J. Cutting edge: contactmediated
suppression by CD4+CD25+ regulatory cells involves
a granzyme Bdependent, perforinindependent
mechanism. J. Immunol. 174, 17831786 (2005).
105. Cao, X. et al. Granzyme B and perforin are important
for regulatory T cellmediated suppression of tumor
clearance. Immunity 27, 635646 (2007).
106. Grossman, W. J. et al. Differential expression of
granzymes A and B in human cytotoxic lymphocyte
subsets and T regulatory cells. Blood 104, 28402848
(2004).
107. Strauss, L., Bergmann, C. & Whiteside, T. L. Human
circulating CD4+CD25highFoxp3+ regulatory T cells
kill autologous CD8+ but not CD4+ responder cells
by Fasmediated apoptosis. J. Immunol. 182,
14691480 (2009).
108. Chen, W. et al. Conversion of peripheral CD4+CD25
naive T cells to CD4+CD25+ regulatory T cells by
TGF induction of transcription factor Foxp3.
J. Exp. Med. 198, 18751886 (2003).

500 | juLy 2010 | VOLuMe 10

109. Andersson, J. et al. CD4+ FoxP3+ regulatory T cells


confer infectious tolerance in a TGFdependent
manner. J. Exp. Med. 205, 19751981 (2008).
110. Belkaid, Y. Regulatory T cells and infection: a
dangerous necessity. Nature Rev. Immunol.
7, 875888 (2007).
111. Roncarolo, M. G. et al. Interleukin10secreting type 1
regulatory T cells in rodents and humans. Immunol.
Rev. 212, 2850 (2006).
112. Garin, M. I. et al. Galectin1: a key effector of
regulation mediated by CD4+CD25+ T cells.
Blood 109, 20582065 (2007).
113. Pandiyan, P., Zheng, L., Ishihara, S., Reed, J. &
Lenardo, M. J. CD4+CD25+Foxp3+ regulatory
T cells induce cytokine deprivationmediated
apoptosis of effector CD4+ T cells. Nature Immunol.
8, 13531362 (2007).
114. Collison, L. W. et al. The inhibitory cytokine IL35
contributes to regulatory Tcell function. Nature 450,
566569 (2007).
115. Bardel, E., Larousserie, F., CharlotRabiega, P.,
CoulombLHermine, A. & Devergne, O. Human
CD4+ CD25+ Foxp3+ regulatory T cells do not
constitutively express IL35. J. Immunol. 181,
68986905 (2008).
116. Gershon, R. K. & Kondo, K. Cell interactions in the
induction of tolerance: the role of thymic lymphocytes.
Immunology 18, 723737 (1970).
117. Sakaguchi, S., Fukuma, K., Kuribayashi, K. &
Masuda, T. Organspecific autoimmune diseases
induced in mice by elimination of T cell subset. I.
Evidence for the active participation of T cells in
natural selftolerance; deficit of a T cell subset as a
possible cause of autoimmune disease. J. Exp. Med.
161, 7287 (1985).
118. Powrie, F. & Mason, D. OX22high CD4+ T cells induce
wasting disease with multiple organ pathology:
prevention by the OX22low subset. J. Exp. Med. 172,
17011708 (1990).
119. Takahashi, T. et al. Immunologic selftolerance
maintained by CD25+CD4+ naturally anergic and
suppressive T cells: induction of autoimmune
disease by breaking their anergic/suppressive state.
Int. Immunol. 10, 19691980 (1998).
120. Nishizuka, Y. & Sakakura, T. Thymus and
reproduction: sexlinked dysgenesia of the gonad
after neonatal thymectomy in mice. Science 166,
753755 (1969).
121. Danke, N. A., Koelle, D. M., Yee, C., Beheray, S. &
Kwok, W. W. Autoreactive T cells in healthy
individuals. J. Immunol. 172, 59675972 (2004).
122. Ghiringhelli, F., Larmonier, N., Schmitt, E. et al.
CD4+CD25+ regulatory T cells suppress tumor
immunity but are sensitive to cyclophosphamide
which allows immunotherapy of established tumors
to be curative. Eur. J. Immunol. 34, 336344 (2004).

Acknowledgements

M.M. is currently supported by Fondation pour la Recherche


Medicale and by the Japan Society for the Promotion of
Science. This work was supported by grantsinaid from the
Ministry of Education, Sports and Culture of Japan, by US
N I H g ra n t s : U O 1 D K 61 9 2 6 01 , R O 1 N S 2 4 2 4 710 ,
PO1AI39671 and PO1NS38037 and grants from the
National Multiple Sclerosis Society: RG2172C9 and
RG3308A10. M.M.

Competing interests statement.

The authors declare no competing financial interests.

DATABASES
UniProtKB: http://www.uniprot.org
CD25 | CD31 | CD62L | CD95 | CD127 | CTLA4 | FOXP3 | GITR |
ICOS | IFN | IL-1 | IL-2 | IL-6 | IL-7 | IL-10 | TSLP
All lInKS ARe AcTIve In THe onlIne PdF

www.nature.com/reviews/immunol
2010 Macmillan Publishers Limited. All rights reserved

Anda mungkin juga menyukai