ArticlePDF Available

Interleukin 7 and T cell receptor signals regulate homeostasis of CD4 memory cells

Authors:
  • Institute of Immunity and Transplantation

Abstract

Immunological memory depends on the long-term maintenance of memory T cells. Although the factors that maintain CD8 T cell memory are well understood, those responsible for CD4 memory are not well defined. We have shown here that interleukin 7 (IL-7) was an important survival factor for CD4 memory T cells that together with T cell receptor (TCR) signals regulated homeostasis of the CD4 memory population in lymphopenic conditions and in the intact immune system. Thus, IL-7 contributes to the maintenance of all naive and memory T cell subsets, and therefore controls the overall size of the T cell pool.
ARTICLES
680 VOLUME 4 NUMBER 7 JULY 2003 NATURE IMMUNOLOGY
The size of the peripheral T cell pool is notably constant despite
thymic production of new T cells and expansion of existing cells after
their stimulation by cognate antigen during immune responses
1
. This
is achieved through specific homeostatic mechanisms that regulate
both cell survival and proliferation. The precise nature of the environ-
mental cues that regulate these responses vary depending on the T cell
subset and on whether the cell is a naive or memory cell. It has been
proposed that these different subsets occupy independently regulated
niches
1
that are maintained by differences in the survival and prolifer-
ative requirements between compartments in both replete and lym-
phopenic conditions.
The homeostatic resources required by naive T cells and CD8
+
memory cells are well defined. The survival of CD4
+
and CD8
+
naive T
cells depends on the cytokine interleukin 7 (IL-7)
2–5
and also requires
contact with self-peptide major histocompatibility complexes
(MHCs)
6–11
. In conditions of lymphopenia, these same ligands drive
naive T cells into homeostatic proliferation
9,12–14
. Memory CD8
+
cells
persist in the absence of class I MHC ligands
15
but require IL-15 for
homeostatic proliferation in replete hosts
16–18
. Although there is no
absolute requirement for IL-7 to maintain CD8 memory cells in
replete mice, IL-7 does contribute to their homeostatic proliferation in
lymphopenic hosts
19,20
and can compensate for the absence of IL-15
(ref. 21). In addition, IL-7 and IL-15 promote survival of CD8 mem-
ory cells in vitro
21
. In contrast, the factors that regulate the survival
and proliferation of memory CD4
+
cells are less well defined and are
instead elucidated by intermittent reports of what is not required.
CD4
+
T cell memory can be generated from effector cells and persist in
the absence of MHC antigens
22
and does not require expression of
either TCR
23
or the Src-family kinases p56
lck
(Lck) and p59
fyn
(Fyn)
for long-term survival
5
. Homeostatic proliferation of CD4 memory
cells in response to lymphopenia occurs in the absence of IL-7 (ref.
24). Furthermore, a study of class II MHC–restricted TCR transgenic
mice lacking expression of the common cytokine receptor γ-chain (γ
c
)
indicates that although naive T cell survival is fundamentally
impaired, activation and memory cell formation is essentially normal,
potentially excluding involvement of all the cytokines whose receptors
require the γ
c
component: IL-2, IL-4, IL-7, IL-9 and IL-15 (ref. 25). It
has therefore been suggested that CD4 memory cells do not require
any specific signals for their homeostatic maintenance
26
.
Despite the burgeoning interest in the signals that influence home-
ostasis of the different T cell subsets, little is known about how CD4
+
memory T cells are regulated. As CD4 memory cells express IL-7
receptor (IL-7R) and receive the same MHC-derived TCR signals that
regulate naive T cell survival in vivo
27
, we sought to determine whether
these pathways were involved in their homeostasis. Using mice
expressing an inducible Lck transgene that permit the manipulation of
signals through the TCR, we were able to dissect the influence of TCR
from IL-7R signals. Our data showed that both of these signals regu-
lated memory CD4 T cell homeostasis. Moreover, we found an overlap
between these signals, such that the influence of one was able to mask
the effects of the other. Therefore, unlike CD8 T cells, naive and mem-
ory CD4 cells were regulated by overlapping signals, challenging the
idea that all T cell subsets necessarily occupy independent niches.
RESULTS
CD4 memory persists without TCR signals
Initially we examined CD4 memory homeostasis in the absence of
TCR stimulation. We used a system that generated resting memory
cells in which TCR signaling could be manipulated. Mice expressing a
tetracycline-regulated inducible Lck transgene (Lck1
ind
; Methods) on
either an Lck
–/–
or Lck
–/–
Fyn
–/–
background reconstitute the T cell
compartment
28
and generate a polyclonal repertoire of naive and
memory (CD44
lo
and CD44
hi
) T cells, similar to that found in wild-
type mice. The wild-type mice and doxycycline-fed Lck1 transgenic
mice (Lck
ind
) used in this study were not deliberately immunized with
antigen, but they nevertheless had T cells with the characteristics of
Division of Molecular Immunology, National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK. Correspondence should be addressed to
R.Z. (rzamoys@nimr.mrc.ac.uk).
Interleukin 7 and T cell receptor signals regulate
homeostasis of CD4 memory cells
Benedict Seddon, Peter Tomlinson & Rose Zamoyska
Immunological memory depends on the long-term maintenance of memory T cells. Although the factors that maintain CD8 T cell
memory are well understood, those responsible for CD4 memory are not well defined. We have shown here that interleukin 7 (IL-
7) was an important survival factor for CD4 memory T cells that together with T cell receptor (TCR) signals regulated homeostasis
of the CD4 memory population in lymphopenic conditions and in the intact immune system. Thus, IL-7 contributes to the
maintenance of all naive and memory T cell subsets, and therefore controls the overall size of the T cell pool.
© 2003 Nature Publishing Group http://www.nature.com/natureimmunology
ARTICLES
NATURE IMMUNOLOGY VOLUME 4 NUMBER 7 JULY 2003 681
memory T cells
29
likely to be specific for environmental antigens
found in gut flora and food. These naturally occurring memory cells in
both wild-type and Lck1
ind
transgenic mice bore all the hallmarks of
memory cells that distinguished them from naive T cells
30,31
: pheno-
typically they were CD44
hi
and CD62L
lo
(Fig. 1a); their survival was
independent of TCR signaling (Fig. 1a)
5
; they had the ability to ‘home’
to tissue sites (Supplementary Fig. 1 online); and, like wild-type mem-
ory T cells, they had the ability to rapidly secrete interferon-γ after
short-term restimulation in vitro (Supplementary Fig. 1 online). We
studied these cells rather than monoclonal TCR transgenic memory
cells because they not only represented a diverse memory pool, but
they also provided the most physiologically relevant setting to examine
signals that regulated homeostasis of CD4 memory cells.
When Lck expression was switched off in adult mice by removal of
doxycycline (Lck1
off
Lck
–/–
Fyn
–/–
), the T cells lacked expression of both
Lck and Fyn and were incapable of being stimulated through the TCR
(data not shown). Thymic development is completely aborted after
removal of doxycycline, so no new naive T cells are produced, and in
the absence of Lck and Fyn, existing naive T cells fail to survive
5
. In
contrast, the CD4
+
CD44
hi
compartment undergoes a brief contrac-
tion (this was approximately twofold in both Lck1
ind
Lck
–/–
and
Lck1
ind
Lck
–/–
Fyn
–/–
mice; data not shown). Although loss of some
memory CD4
+
cells may contribute to this reduction, the absence of
Lck will also cause loss of effector cells, absence of new input from the
naive pool and reduced turnover of existing memory cells. The
remaining population of CD4
+
CD44
hi
T cells in these mice persists in
the absence of TCR signaling
5
. Within several weeks, their CD4
+
com-
partment contained almost exclusively CD44
hi
CD62L
lo
memory phe-
notype cells (Fig. 1a), in contrast to that of either wild-type or
doxycycline-fed Lck1
ind
Lck
–/–
Fyn
–/–
mice, in which both naive and
memory T cells were supported. The range of TCR V
β
expression
among the persisting CD4
+
CD44
hi
T cells was similar to that found in
wild-type and doxycycline-fed mice, confirming the polyclonality of
this memory population (Supplementary Fig. 1 online). Because we
wanted to examine the influence of IL-7R signals on CD4 memory cell
behavior in these mice, we confirmed that IL-7R expression was nor-
mal. CD4
+
CD44
hi
-phenotype cells both from wild-type and the vari-
ous Lck1 mice, on or off doxycycline, expressed large amounts of
IL-7R (Fig. 1b). Expression of IL-7R was slightly increased on the
memory population from Lck1
off
Lck
–/–
Fyn
–/–
mice, perhaps because
of the loss of TCR signaling capacity, as signals through the TCR
down-modulate IL-7R expression
2
. These data confirmed that TCR
signals were not required for long-term survival of polyclonal memory
CD4 T cells.
Influence of IL-7 on CD4 memory
Because CD4 memory can persist in the absence of antigen, MHC lig-
ands and Src kinases, we investigated whether IL-7 was involved in
their homeostasis. We obtained CD4
+
CD44
hi
T cells from
Lck1
off
Lck
–/–
Fyn
–/–
mice off doxycycline for between 1 and 3 months,
labeled the cells with carboxyfluorescein diacetate succinimidyl ester
(CFSE) and transferred them to recipient mice deficient in recombina-
tion activating gene 1 (Rag1
–/–
): Il7
+/+
Rag1
–/–
, Il7
+/–
Rag1
–/–
or
Il7
–/–
Rag1
–/–
. On days 1, 21 and 35 after transfer, we assessed T cell
recoveries from lymph nodes and spleens of recipient mice and exam-
ined CFSE labeling of donor cells. After being transferred to
Il7
+/+
Rag1
–/–
recipients, Lck1
off
Lck
–/–
Fyn
–/–
memory CD4
+
cells
underwent a number of divisions that increased over time (Fig. 2a).
Moreover, cell proliferation was also accompanied by an increase in
cell recovery. Cells transferred to Il7
–/–
Rag1
–/–
recipients failed to
divide. Although recoveries from Il7
+/+
Rag1
–/–
and Il7
–/–
Rag1
–/–
mice
were identical at day 1 after cell transfer (Fig. 2b), indicating that the
initial seeding of transferred cells was similar between the recipient
strains, on days 21 and 35 after transfer, cell recoveries were far greater
from mice expressing IL-7 than from Il7
–/–
Rag1
–/–
recipients (Fig. 2b).
To determine whether IL-7 was influencing both expansion and
Figure 1 Persistence of memory CD4 T cells in the absence of TCR signaling. Lck1
ind
Lck
–/–
Fyn
–/–
mice were switched from doxycycline-containing food
to conventional diets to turn off Lck expression (Lck1
off
Lck
–/–
Fyn
–/–
). Lymph node cells from these mice or Lck1
ind
Lck
–/–
Fyn
–/–
mice fed doxycycline
continuously and from wild-type controls were stained for CD4, CD44, CD62L and TCR and were analyzed by FACS. (a) CD4 versus CD8 expression on live-
gated lymph node cells (top row) and CD62L versus CD44 expression on gated CD4
+
TCR
hi
lymph node cells (bottom row) from wild-type controls (left
column), doxycycline-fed Lck1
ind
Lck
–/–
Fyn
–/–
mice (middle column) and Lck1
off
Lck
–/–
Fyn
–/–
mice (off doxycycline for 6 weeks; right column). Percentages
indicate frequencies of CD4
+
and CD8
+
cells (top row) or frequencies of CD62L
hi
CD44
lo
and CD62L
lo
CD44
hi
populations (bottom row). (b) IL-7R expression
by either CD4
+
CD44
hi
or CD4
+
CD44
lo
cells from age-matched wild-type, Lck1
ind
Lck
–/–
or Lck1
ind
Lck
–/–
Fyn
–/–
mice either maintained on or taken off the
doxycycline-containing diet for at least 2 weeks. Vertical markers were set against negative staining controls and indicate the threshold of positive staining.
+ Dox, with doxycycline; – Dox, without doxycycline.
ab
© 2003 Nature Publishing Group http://www.nature.com/natureimmunology
ARTICLES
682 VOLUME 4 NUMBER 7 JULY 2003 NATURE IMMUNOLOGY
survival of the transferred cells, we adjusted results for cell recoveries
to compensate for the effects of cell division (Fig. 2c). Even with this
adjustment, cell recoveries were approximately three times greater
from Il7
+/+
Rag1
–/–
mice than from Il7
–/–
Rag1
–/–
recipients at day 21
(Fig. 2c). By day 35, all cells transferred to Il7
+/+
Rag1
–/–
mice had lost
their CFSE labeling, so those results could not be adjusted for division
number; however, the difference in cell recoveries between
Il7
+/–
Rag1
–/–
and Il7
–/–
Rag1
–/–
became greater over time (Fig. 2c).
Examination of peritoneal exudate cells showed that cell recoveries
from the tissues at day 35 (Fig. 2d) mirrored those from lymph node
and spleen, indicating that loss of cells from Il7
–/–
mice was not simply
a reflection of differences in ‘homing’. Inclusion of Il7
+/–
Rag1
–/–
recip-
ients showed a distinct gene-dose effect for IL-7. Cells transferred to
these hosts proliferated less (Fig. 2a), accumulated less (Fig. 2b) and
survived less well (Fig. 2c) than did cells in Il7
+/+
Rag1
–/–
hosts, con-
firming that the availability of IL-7 directly influenced both expansion
and survival of CD4
+
memory cells.
CD4 memory maintenance without IL-7R signals
Our data demonstrated involvement of IL-7 in the homeostasis of
CD4 memory, but previous studies have shown that mice with
impaired cytokine signaling generate CD4 memory cells that per-
sist
32–34
. Because we showed that in the absence of both TCR and IL-
7R signals, CD4 memory T cells could not proliferate and failed to
survive, we asked what function TCR signals have in the homeostasis
of CD4 memory.
We first examined the phenotype of IL-7R-deficient mice, which
are profoundly lymphopenic due to a developmental block in T and
B cell development caused by the absence of this receptor
35,36
.
However, analysis of splenocytes from these mice showed that
although naive phenotype CD4
+
CD44
lo
CD62L
hi
cells were scarce,
memory phenotype CD4
+
CD44
hi
CD62L
lo
cells were unexpectedly
abundant (Fig. 3a), as in other mice with defective cytokine signal-
ing
32–34
. Therefore, CD4
+
CD44
hi
memory phenotype cells were
produced in response to TCR signals in these mice, in numbers simi-
lar to those of wild-type mice (Fig. 3b).
ab
c
d
Figure 3 IL-7R signals were not required for the generation of CD4
+
CD44
hi
cells. Spleen cells from Il7r
–/–
and wild-type control mice (n = 5) were stained
for CD4, CD8, CD44, CD62L and TCR expression and analyzed by FACS.
(a) CD4 versus CD8 expression on splenocytes gated on live cells (top row),
and CD62L versus CD44 expression by gated CD4
+
TCR
+
splenocytes from
wild-type and Il7r
–/–
mice (bottom row). Percentages indicate frequencies of
CD4
+
cells (top row) or CD62L
hi
CD44
lo
and CD62L
lo
CD44
hi
populations
(bottom row). (b) Total numbers of CD4
+
CD44
lo
and CD4
+
CD44
hi
cells
recovered from the mice in a.
ab
Figure 2 IL-7R signals promoted proliferation and survival of CD4
+
CD44
hi
cells in the absence of TCR signaling. CD4
+
CD44
hi
cells, purified from
Lck1
ind
Lck
–/–
Fyn
–/–
mice taken off doxycycline 4–12 weeks previously,
were labeled with CFSE and transferred to Il7
+/+
Rag1
–/–
, Il7
+/–
Rag1
–/–
or
Il7
–/–
Rag1
–/–
recipients (n =3 per group). (a) CFSE profiles of gated
CD4
+
TCR
hi
CD44
hi
cells from blood (day 6) or spleen (days 21 and 35).
(b,c) Total numbers of CD4
+
CD44
hi
TCR
+
cells recovered from lymph nodes
and spleens from different recipient mice (b) or adjusted figures that
compensated for the effects of cell division on cell numbers (c). The expansion
index was calculated from CFSE profiles (Methods) as 3.9 and 1.75 for cells from Il7
+/+
Rag1
–/–
and Il7
+/–
Rag1
–/–
hosts, respectively, on day 21, and 6.7 for cells
from Il7
+/–
Rag1
–/–
hosts on day 35. N.D., not determined. (d) CD4 and TCR expression by lymphocytes recovered from the peritoneum of recipient mice 35 days
after cell transfer. Percentages indicate frequencies of CD4
+
TCR
hi
cells. Data were pooled from three independent experiments in which mice received either
1 × 10
6
(day 21) or 4 × 10
6
(days 1 and 35) memory cells per mouse and were representative of two further experiments.
© 2003 Nature Publishing Group http://www.nature.com/natureimmunology
ARTICLES
NATURE IMMUNOLOGY VOLUME 4 NUMBER 7 JULY 2003 683
The fact that thymic selection in IL-7R-deficient mice and other mice
lacking essential cytokine signaling components
25,32–34
is profoundly
blocked may favor selection of T cells that are relatively cytokine-
independent. Therefore, we also examined the ability of wild-type
CD4
+
CD44
hi
cells to undergo homeostatic responses after their trans-
fer to lymphopenic hosts in the absence of IL-7. CFSE labeled
CD4
+
CD44
hi
cells from wild-type mice were transferred into
Il7
+/+
Rag1
–/–
, Il7
+/–
Rag
–/–
, Il7
–/–
Rag1
–/–
recipient mice or Rag2
–/–
recipient mice deficient in the IL-2 receptor common γ-chain
(Il2rg
–/–
Rag2
–/–
). Il2rg
–/–
Rag2
–/–
mice have the same defects in lym-
phoid architecture as do Il7
–/–
Rag1
–/–
mice
37
, lacking almost all lymph
nodes
33
, but still express IL-7. As with Lck1
ind
Lck
–/–
Fyn
–/–
T cells trans-
ferred to Il7
+/+
Rag1
–/–
or Il7
–/–
Rag1
–/–
hosts (Fig. 2), recoveries of
wild-type T cells from all these hosts were similar at day 1 (data not
shown). By day 7, analysis of peripheral blood showed that transferred
cells had undergone extensive proliferation, losing CFSE dye, regardless
of whether hosts had IL-7 or not (Fig. 4a), as reported before
19
.
Similarly, transfer of these cells to Rag1
–/–
hosts treated with antibody
to IL-7R had little effect on the proliferative response of the cells
(Fig. 4b). However, donor cell numbers at day 14 showed that in the
absence of IL-7, or in the presence of IL-7R-blocking antibodies, there
was a profound reduction in cell recovery (Fig. 4c). This reduction
could not be attributed to the altered state of the lymphoid compart-
ment in Il7
–/–
Rag1
–/–
mice, as recoveries from Il2rg
–/–
Rag2
–/–
hosts,
which have a similar lymphoid defect, were normal. In addition, we
found a similar reduction after treatment of normal Rag1
–/–
recipients
Figure 4 Proliferation but no accumulation of
wild-type CD4
+
CD44
hi
T cells in the absence
of IL-7. CD4
+
CD44
hi
cells were purified from
spleens of 6- to 8-week-old wild-type mice,
labeled with CFSE and transferred (4 × 10
6
cells
per mouse) to Rag1
–/–
recipient mice that were
Il7
+/+
, Il7
+/–
or Il7
–/–
, and to Il2rg
–/–
Rag2
–/–
recipient mice. (a) On days 7 and 14, blood
was obtained from mice, and CFSE labeling
of CD4
+
CD44
hi
cells in peripheral blood was
determined by staining of cells for CD4, TCR
and CD44 expression and analysis by FACS.
(b) Using a protocol similar to that in a.
CD4
+
CD44
hi
cells from wild-type mice were
transferred to Rag1
–/–
recipients (4 × 10
6
cells
per mouse) treated with monoclonal antibody to
IL-7R or with PBS as a control. CFSE labeling in
peripheral blood was measured on days 7 and 14.
(c) Total numbers of CD4
+
TCR
+
cells recovered
from lymph nodes and spleens of at least three
recipient mice per group described in a and b.
Data were representative of two independent
experiments.
ab
c
ab
c
de
Figure 5 Maintenance of CD4 memory in full mice by TCR signals and IL-7.
(a) CD44 versus antibody-to-BrdU staining by CD4
+
TCR
+
peripheral blood
lymphocytes from wild-type mice and doxycycline-fed Lck1
ind
Lck
–/–
and
Lck1
off
Lck
–/–
Fyn
–/–
mice (n =3 per group) fed BrdU-containing water for
1 week. Percentages indicate the mean frequency ± s.d. of CD4
+
CD44
hi
cells
staining for BrdU incorporation. (be) Lck1
ind
Lck
–/–
mice 6–8 weeks of age
were taken off doxycycline and treated either with monoclonal antibody to
IL-7R or with PBS as control. At 0, 2 and 4 weeks after treatment, total
numbers of CD8
+
CD44
hi
and CD4
+
CD44
hi
cells from groups of mice (n = 3)
were determined by counting of total cells in lymph nodes and analysis of the
expression of CD8, CD4, TCR and CD44 by FACS. Data represent average
numbers ± s.d. of recovered CD4
+
CD44
lo
(b), CD4
+
CD44
hi
(c), CD8
+
CD44
lo
(d) and CD8
+
CD44
hi
(e) cells in mice treated with antibody to IL-7R (open
symbols) or control mice (filled symbols). Data were representative of three
(a) or four (be) independent experiments.
© 2003 Nature Publishing Group http://www.nature.com/natureimmunology
ARTICLES
684 VOLUME 4 NUMBER 7 JULY 2003 NATURE IMMUNOLOGY
with antibody to IL-7R. These data confirmed that TCR-mediated sig-
nals were able to induce proliferation of CD4
+
memory T cells and,
therefore, contributed to their persistence in the absence of IL-7. The
presence of CD4 memory cells in Il7r
–/–
mice and the observation that
some wild-type CD4 memory cells were recovered after transfer to
Il7
–/–
Rag1
–/–
hosts indicated limited involvement of TCR signals in
promoting survival of these cells. However, the decrease in recovery of
cells in the absence of IL-7 showed an important function for this
cytokine in CD4 memory cell survival.
CD4 memory cells in replete hosts
Having established the signals that regulated CD4 memory T cell
homeostasis in lymphopenic conditions, we asked how these signals
contributed to the maintenance of these cells in normal mice in
steady-state conditions. Memory cells undergo nonexpansive cell divi-
sions in replete hosts
38
and this slow turnover may contribute to their
ability to mount more rapid responses than naive cells, by allowing
quicker entry into cell cycle. Although TCR signals do not contribute
much to the long-term survival of CD4 memory cells, we asked
whether these signals were responsible for the memory cell turnover in
replete mice. We fed bromodeoxyuridine (BrdU) to wild-type mice or
control doxycycline-fed Lck1
ind
Lck
–/–
and Lck1
off
Lck
–/–
Fyn
–/–
mice for
1 week and analyzed T cells for BrdU incorporation. Although naive T
cells did not incorporate any BrdU, just over 50% of CD4
+
CD44
hi
cells
from Lck1
ind
Lck
–/–
mice and wild-type controls had undergone cell
division in 1 week (Fig. 5a). In contrast, less than 10% of CD4
+
CD44
hi
cells from Lck1
off
Lck
–/–
Fyn
–/–
mice had incorporated BrdU during this
time (Fig. 5a), indicating that TCR signals were important in stimulat-
ing proliferation of the CD4 memory compartment of replete mice.
Although we were able to show that TCR signals induced prolifer-
ation of CD4 memory T cells in replete mice, neither proliferation
(Fig. 5a) nor TCR signals
5
were essential for the long-term survival
of these cells. We asked, therefore, whether IL-7 was important for
the maintenance of CD4 memory in unmanipulated replete hosts.
We examined the survival of memory T cells in mice treated with a
blocking antibody to IL-7R. In these experiments, we used
Lck1
ind
Lck
–/–
mice taken off doxycycline, to maintain a static T cell
pool. In the absence of Lck expression not only does thymic atrophy
prevent export of newly differentiated T cells, but also the incum-
bent peripheral T cells are unable to expand to TCR-mediated
homeostatic signals
39
. However, both naive and memory cells still
survive in the absence of Lck alone. We treated groups of
Lck1
off
Lck
–/–
mice with antibody to IL-7R or with PBS for 4 weeks.
We determined naive and memory T cell subset numbers in lymph
nodes obtained from mice 0, 2 and 4 weeks after treatment was
begun. As reported before
5
, blockade of IL-7R resulted in loss of
both naive CD4
+
CD44
lo
(Fig. 5b) and CD8
+
CD44
lo
(Fig. 5d) T cells.
In the presence of IL-7R blockade, CD4
+
CD44
hi
cells (Fig. 5c)
showed a decrease in number similar to that of their naive counter-
parts. There was also an initial smaller decrease in CD4
+
CD44
hi
cells
in the PBS-treated control mice, which indicated that there was a
subset of these cells that relied on TCR-mediated signals for their
persistence. Consistent with published results
20
, CD8
+
CD44
hi
cell
numbers were completely unaffected by treatment with antibody to
IL-7R (Fig. 5e), indicating that the antibody did not eliminate T
cells by antibody-dependent cell-mediated cytotoxicity in vivo, as
CD8 memory cells also expressed large amounts of IL-7R
(Supplementary Fig. 1 online). These results confirmed that
although TCR-mediated signals could contribute to turnover of
CD4 memory cells in an intact mouse, the maintenance of CD4
memory cell numbers depended on the availability of IL-7.
DISCUSSION
The signals that regulate CD4 memory cell homeostasis are ill-defined.
In this study, we examined whether signaling by the IL-7R and TCR
contributed to the survival and homeostatic proliferation of CD4
memory cells in normal mice. Our data showed that in conditions of
lymphopenia and in the intact immune system, CD4 memory cell
homeostasis was regulated through the combined effects of TCR and
IL-7R signaling. In the absence of both these signals, CD4 memory
cells were unable to divide and failed to survive.
Our data seemed to contradict published reports indicating no
involvement of the TCR
22,23
or γ
c
cytokines
19,25
in the maintenance
of CD4 memory cells. One possible explanation for this discrepancy
is that TCR and IL-7R signals can have similar outcomes with
respect to the maintenance of CD4 memory cell numbers. We
showed here that in the absence of TCR signals, a polyclonal popula-
tion of CD4 memory cells survived long term and turned over
slowly in response to IL-7. However, mice with defective cytokine
signaling maintained a population of memory phenotype cells sim-
ilar in size to that of wild-type mice. CD4 memory cell persistence in
mice deficient in cytokine signaling will be influenced by extensive
lymphopenia and/or environmental antigen-induced cell division
which may mask defects in cell survival. Indeed, polyclonal Jak3
–/–
(ref. 40) and Il2rg
–/–
(ref. 34) mice have more extensive proliferation
(assessed by BrdU uptake) of CD4
+
CD44
hi
cells than do wild-type
mice, and have excessive cell death. Furthermore, spectratype analy-
sis of memory cells from Jak3
–/–
mice shows a skewed TCR reper-
toire
40
, indicating the selection of an autoantigen-reactive,
TCR-driven population, and these mice ultimately develop colitis.
These data also indicate that TCR and IL-7 signals are not acting
redundantly, as they have different outcomes on the composition of
the memory population. TCR signals may preferentially promote
survival of cells proliferating in response to persistent antigen,
whereas IL-7 may promote survival of memory cells that do not
require TCR signals and therefore do not require the presence of
antigen. In normal mice with full T cell compartments, there is
clearly extensive TCR-driven cell division in the memory pool, but
no accompanying expansion. In such situations, competition for IL-
7 most likely regulates the overall pool size. It is possible that cell
division induced by TCR stimulation fulfills a different function,
perhaps being more important for the functional competence of the
cell. Indeed, although memory cells may persist in the absence of
MHC ligands, they do show considerable functional defects
41
.
Our findings also indicated that the signals required for homeostasis
of CD4 memory were essentially the same as those that regulated naive
CD4
+
T cells: IL-7 and TCR signals. The fact that naive and memory
cells shared common resources seemed initially at odds with the view
that these populations should occupy non-overlapping and non-com-
peting niches
1
. Although the behavior of the naive and memory com-
partments may be operationally independent, there are examples of
CD4 memory phenotype cells interfering with the homeostasis of
naive T cells. For example, the few T cells present in Lck
–/–
Fyn
+/–
mice,
all of which are of a memory phenotype, can prevent almost all IL-7-
driven proliferation of naive T cells
42
, presumably by competing for
available IL-7. However, the key difference between the homeostasis of
naive and memory CD4 cells may be the degree of redundancy
between the signals involved, rather than the nature of the specific sur-
vival factors. Thus, memory CD4 cells are substantially influenced by
either IL-7R or TCR signaling, whereas naive CD4 cells require the co-
operative activity of both of these receptor signaling pathways, and this
operational difference in the use of common resources allows their rel-
atively independent regulation.
© 2003 Nature Publishing Group http://www.nature.com/natureimmunology
ARTICLES
NATURE IMMUNOLOGY VOLUME 4 NUMBER 7 JULY 2003 685
Our finding that IL-7 was important in the maintenance of CD4
memory identifies a single factor that regulates the homeostasis of all
naive and memory peripheral T cell subsets. Although there are subtle
differences in the specific requirements and use of resources by these
subsets that may afford a degree of independence, the universal
requirement of IL-7 strongly indicates it is the master regulator that
controls the overall size of the T cell compartment.
METHODS
Mice. The generation of inducible Lck transgenic Lck1
ind
mice has been
described
28
. Mice expressing the reverse tetracycline-responsive transactivator
domain (rtTA) under control of the human CD2 promotor on an endogenous
Lck
–/–
background (rtTA-C Lck
–/–
) were intercrossed with mice bearing the
mouse Lck transgene under control of a tetO-CMVmin promotor, also on an
endogenous Lck
–/–
background (Lck1Lck
–/–
). Mice were given doxycycline in
food (1 mg per g body weight) throughout pregnancy and before weaning.
After being weaned, Lck1 rtTA-C Lck
–/–
F1 progeny were given food containing
doxycycline to maintain Lck1 transgene expression (Lck1
ind
Lck
–/–
) or were
switched to conventional diets to shut off transgene expression (Lck1
off
Lck
–/–
).
Lck1
ind
Lck
–/–
Fyn
–/–
mice were similarly derived by intercrossing rtTA-C
Lck
–/–
Fyn
–/–
and Lck1Lck
–/–
Fyn
–/–
strains. C57BL/10 wild-type, Rag1
–/–
, Il7r
–/–
,
Il7
–/–
Rag1
–/–
and Il2rg
–/–
Rag2
–/–
mice were bred in a conventional colony free of
pathogens at National Institute for Medical Research (London, UK). All lines
used were of H-2
b
haplotype. Animal experiments were done according to
institutional guidelines and Home Office regulations.
Flow cytometry and cellularity determinations. Flow cytometry used 2 × 10
6
to 5 × 10
6
lymph node, spleen cells or peritoneal exudate cells. Lymph node
cell suspensions were prepared from superficial cervical, brachial, axillary,
inguinal and mesenteric lymph nodes of individual mice, whereas peritoneal
exudate cells were collected after injection of 5 ml media into the peritoneal
cavities of mice. Cell concentrations were determined using a Scharf
Instruments Casy Counter. Cells were incubated for 1 h at 4 °C with saturating
concentrations of antibodies in 100 µl PBS in BSA (0.1%) plus azide (1 mM),
followed by a three washes in the buffer of PBS, BSA and azide. Biotin-
conjugated antibody labeling was developed with streptavidin–peridinin
chlorophyll protein (PharMingen). Monoclonal antibodies used in this study
were: allophycocyanin- and phycoerythrin-conjugated CD4 (GK1.5;
PharMingen); fluorescein isothiocyanate–conjugated TCR (H57-597;
PharMingen); allophycocyanin-conjugated CD44 (Leinco Technologies);
phycoerythrin-conjugated CD44 (PharMingen); Mel14 (antibody to CD62L);
allophycocyanin-conjugated CD8 (PharMingen); and IL-7 receptor α-chain
(A7R34). Four-color cytometric staining was analyzed on a FACSCalibur
Instrument (Becton Dickinson) and data were analyzed with Cell Quest soft-
ware (Becton Dickinson). To determine cell proliferation in vivo by BrdU
incorporation, mice were first injected intraperitoneally with 1 mg BrdU, then
were fed BrdU in drinking water (0.8 mg/ml) protected from light. BrdU water
was renewed every 2 d. Intracellular staining for BrdU incorporation and
cytokines was done using Fastimmune Anti-BrdU with DNase (Becton
Dickinson) and antibody to interferon-g (PharMingen), respectively, accord-
ing to manufacturers’ instructions.
Purification, labeling and adoptive transfer of T cells. Lymphocytes were
‘teased’ from lymph nodes and spleens of donor mice, and single-cell suspen-
sions were prepared. For experiments requiring purified CD4
+
CD44
hi
T cell
preparations, lymphocytes were labeled for 30 min at 4 °C with a mixture of
antibodies to CD62L-biotin and DX5-biotin (PharMingen), CD8-biotin and
B220-biotin (PharMingen) and I-A
b
-biotin (PharMingen) at a concentration
of 1 µg/ml (100 µl per 1 × 10
7
cells). After being washed, cells were incubated
for 30 min with M280-streptavidin Dynal beads (Dynal) , after which cells were
separated by application of a magnet. Contaminating cells were consistently
<1%. Cells were labeled for 10 min at 37 °C with 2 µM CFSE (Molecular
Probes) in Dulbeccos PBS (Life Technologies) and washed twice. Cells were
transferred into various recipient mice through tail vein injections. Treatment
of recipient mice with purified monoclonal antibody to IL-7R (A7R34; a gift
from S. Nishikawa, Kyoto University, Kyoto, Japan) in PBS was done on a 7-day
cycle of intraperitoneal injections of 500 µg/mouse of monoclonal antibody on
days 1, 3 and 5 for the duration of the experiment. Control mice received injec-
tions of PBS alone. Cellular expansion of CFSE-labeled cells was calculated by
determining the frequency (F) of donor cells that had undergone different
numbers of divisions (d). Adjusted frequencies (AdF) were calculated by divid-
ing F by 2
d
. The predicted expansion was determined by the calculation
ΣF/ΣAdF.
Note: Supplementary information is available on the Nature Immunology website.
ACKNOWLEDGMENTS
We thank T. Norton, K. Williams and the Biological Services staff for assistance with
mouse breeding and typing. We also thank B. Stockinger and G. Kassiotis, as well as
other members of the Division of Molecular Immunology, for discussions. This
work was supported by the Medical Research Council and the Leukaemia Research
Fund, UK.
COMPETING INTERESTS STATEMENT
The authors declare that they have no competing financial interests.
Received 13 March; accepted 28 May 2003
Published online 15 June 2003; doi:10.1038/ni946
1. Freitas, A.A. & Rocha, B. Population biology of lymphocytes: the flight for survival.
Annu. Rev. Immunol. 18, 83–111 (2000).
2. Schluns, K.S., Kieper, W.C., Jameson, S.C. & Lefrancois, L. Interleukin-7 mediates
the homeostasis of naive and memory CD8 T cells in vivo. Nat. Immunol. 1,
426–432 (2000).
3. Vivien, L., Benoist, C. & Mathis, D. T lymphocytes need IL-7 but not IL-4 or IL-6 to
survive in vivo. Int. Immunol. 13, 763–768 (2001).
4. Tan, J.T. et al. IL-7 is critical for homeostatic proliferation and survival of naive T
cells. Proc. Nat. Acad. Sci. USA 98, 8732–8737 (2001).
5. Seddon, B. & Zamoyska, R. TCR signals mediated by Src family kinases are essential
for the survival of naive T cells. J. Immunol. 169, 2997–3005 (2002).
6. Tanchot, C., Lemonnier, F.A., Perarnau, B., Freitas, A.A. & Rocha, B. Differential
requirements for survival and proliferation of CD8 naive or memory T cells. Science
276, 2057–2062 (1997).
7. Kirberg, J., Berns, A. & von Boehmer, H. Peripheral T cell survival requires continual
ligation of the T cell receptor to major histocompatibility complex-encoded mole-
cules. J. Exp. Med. 186, 1269–1275 (1997).
8. Brocker, T. Survival of mature CD4 T lymphocytes is dependent on major histocom-
patibility complex class II-expressing dendritic cells. J. Exp. Med. 186,
1223–1232 (1997).
9. Viret, C., Wong, F.S. & Janeway, C.A., Jr. Designing and maintaining the mature TCR
repertoire: the continuum of self-peptide:self-MHC complex recognition. Immunity
10, 559–568 (1999).
10. Rooke, R., Waltzinger, C., Benoist, C. & Mathis, D. Targeted complementation of
MHC class II deficiency by intrathymic delivery of recombinant adenoviruses.
Immunity 7, 123–134 (1997).
11. Witherden, D. et al. Tetracycline-controllable selection of CD4
+
T cells: half-life and
survival signals in the absence of major histocompatibility complex class II mole-
cules. J. Exp. Med. 191, 355–364 (2000).
12. Goldrath, A.W. & Bevan, M.J. Low-affinity ligands for the TCR drive proliferation of
mature CD8
+
T cells in lymphopenic hosts. Immunity 11, 183–190 (1999).
13. Kieper, W.C. & Jameson, S.C. Homeostatic expansion and phenotypic conversion of
naive T cells in response to self peptide/MHC ligands. Proc. Natl. Acad. Sci. USA 96,
13306–13311 (1999).
14. Ernst, B., Lee, D.S., Chang, J.M., Sprent, J. & Surh, C.D. The peptide ligands medi-
ating positive selection in the thymus control T cell survival and homeostatic prolifer-
ation in the periphery. Immunity 11, 173–181 (1999).
15. Murali-Krishna, K. et al. Persistence of memory CD8 T cells in MHC class I-deficient
mice. Science 286, 1377–1381 (1999).
16. Zhang, X., Sun, S., Hwang, I., Tough, D.F. & Sprent, J. Potent and selective stimulation
of memory-phenotype CD8+ T cells in vivo by IL-15. Immunity 8, 591–599 (1998).
17. Ku, C.C., Murakami, M., Sakamoto, A., Kappler, J. & Marrack, P. Control of homeosta-
sis of CD8
+
memory T cells by opposing cytokines. Science 288, 675–678 (2000).
18. Becker, T.C. et al. Interleukin 15 is required for proliferative renewal of virus-specific
memory CD8 T cells. J. Exp. Med. 195, 1541–1548 (2002).
19. Tan, J.T. et al. Interleukin (IL)-15 and IL-7 jointly regulate homeostatic proliferation
of memory phenotype CD8+ cells but are not required for memory phenotype CD4
+
cells. J. Exp. Med. 195, 1523–1532 (2002).
20. Goldrath, A.W. et al. Cytokine requirements for acute and basal homeostatic prolifer-
ation of naive and memory CD8
+
T cells. J. Exp. Med. 195, 1515–1522 (2002).
21. Kieper, W.C. et al. Overexpression of interleukin (IL)-7 leads to IL-15-independent gen-
eration of memory phenotype CD8
+
T cells. J. Exp. Med. 195, 1533–1539 (2002).
22. Swain, S.L., Hu, H. & Huston, G. Class II-independent generation of CD4 memory T
cells from effectors. Science, 1381–1383 (1999).
23. Polic, B., Kunkel, D., Scheffold, A. & Rajewsky, K. How αβ-T cells deal with induced
TCR-α ablation. Proc. Natl. Acad. Sci. USA 98, 8744–8749 (2001).
24. Tan, J.T. et al. Interleukin (IL)-15 and IL-7 jointly regulate homeostatic proliferation
© 2003 Nature Publishing Group http://www.nature.com/natureimmunology
ARTICLES
686 VOLUME 4 NUMBER 7 JULY 2003 NATURE IMMUNOLOGY
of memory phenotype CD8
+
cells but are not required for memory phenotype CD4
+
cells. J. Exp. Med. 195, 1523–1532 (2002).
25. Lantz, O., Grandjean, I., Matzinger, P. & Di Santo, J.P. γ chain required for naive CD4
+
T cell survival but not for antigen proliferation. Nat. Immunol. 1, 54–58 (2000).
26. Jameson, S.C. Maintaining the norm: T-cell homeostasis. Nat. Rev. Immunol. 2,
547–556 (2002).
27. Dorfman, J.R., Stefanova, I., Yasutomo, K. & Germain, R.N. CD4
+
T cell survival is not
directly linked to self-MHC-induced TCR signaling. Nat. Immunol. 1, 329–335 (2000).
28. Legname, G. et al. Inducible expression of a p56Lck transgene reveals a central role
for Lck in the differentiation of CD4 SP thymocytes. Immunity 12, 537–546 (2000).
29. Swanson, B.J., Murakami, M., Mitchell, T.C., Kappler, J. & Marrack, P. RANTES pro-
duction by memory phenotype T cells is controlled by a posttranscriptional, TCR-
dependent process. Immunity 17, 605–615 (2002).
30. Swain, S.L. et al. Helper T-cell subsets: phenotype, function and the role of lym-
phokines in regulating their development. Immunol. Rev. 123, 115–144 (1991).
31. Sprent, J. T and B memory cells. Cell 76, 315–322 (1994).
32. Thomis, D.C., Gurniak, C.B., Tivol, E., Sharpe, A.H. & Berg, L.J. Defects in B lym-
phocyte maturation and T lymphocyte activation in mice lacking Jak3. Science 270,
794–797 (1995).
33. Cao, X. et al. Defective lymphoid development in mice lacking expression of the com-
mon cytokine receptor gamma chain. Immunity 2, 223–238 (1995).
34. Nakajima, H., Shores, E.W., Noguchi, M. & Leonard, W.J. The common cytokine
receptor γ-chain plays an essential role in regulating lymphoid homeostasis. J. Exp.
Med. 185, 189–195 (1997).
35. Peschon, J.J. et al. Early lymphocyte expansion is severely impaired in interleukin 7
receptor-deficient mice. J. Exp. Med. 180, 1955–1960 (1994).
36. Maraskovsky, E. et al. Impaired survival and proliferation in IL-7 receptor-deficient
peripheral T cells. J. Immunol. 157, 5315–5323 (1996).
37. von Freeden-Jeffry, U. et al. Lymphopenia in interleukin (IL)-7 gene-deleted mice
identifies IL-7 as a nonredundant cytokine. J. Exp. Med. 181, 1519-1526 (1995).
38. Tough, D.F. & Sprent, J. Turnover of naive- and memory-phenotype T cells. J. Exp.
Med. 179, 1127–1135 (1994).
39. Seddon, B., Legname, G., Tomlinson, P. & Zamoyska, R. Long-term survival but
impaired homeostatic proliferation of naive T cells in the absence of p56
lck
. Science
290, 127–131 (2000).
40. Gozalo-Sanmillan, S., McNally, J.M., Lin, M.Y., Chambers, C.A. & Berg, L.J. Cutting
edge: two distinct mechanisms lead to impaired T cell homeostasis in Janus kinase 3-
and CTLA-4-deficient mice. J. Immunol. 166, 727–730 (2001).
41. Kassiotis, G., Garcia, S., Simpson, E. & Stockinger, B. Impairment of immunological
memory in the absence of MHC despite survival of memory T cells. Nat. Immunol. 3,
244–250 (2002).
42. Seddon, B. & Zamoyska, R. TCR and IL-7 receptor signals can operate independently
or synergize to promote lymphopenia-induced expansion of naive T cells. J. Immunol.
169, 3752–3759 (2002).
© 2003 Nature Publishing Group http://www.nature.com/natureimmunology
... Transcriptional factor KLF2 regulates the development of CD62L-expressing Tfh cells In the single-cell RNA-seq data, Il7r expression correlated with Sell, but not Pdcd1 expression in Tfh cells (Fig. 1 B). The ATACseq analysis also revealed that the Il7r gene locus was more accessible in CD62L + than PD-1 + Tfh cells (Fig. S5 A), suggesting that the IL7R signaling, an important regulator in memory T cell generation and maintenance (Seddon et al., 2003;Kondrack et al., 2003;Li et al., 2003;Kitano et al., 2011;McDonald et al., 2016), may contribute to the regulation of CD62L + Tfh cells. We therefore administrated anti-IL7R blocking antibody to CD45.1 recipient mice transferred with naïve CD4 + T cells from Bcl6 RFP × Foxp3 GFP OT-II transgenic mice and immunized with OVA (Fig. S5 B). ...
Article
Full-text available
T follicular helper (Tfh) cells, essential for germinal center reactions, are not identical, with different phenotypes reported. Whether, when, and how they generate memory cells is still poorly understood. Here, through single-cell RNA-sequencing analysis of CXCR5⁺Bcl6⁺ Tfh cells generated under different conditions, we discovered, in addition to PD-1hi effector Tfh cells, a CD62L⁺PD1low subpopulation. CD62L-expressing Tfh cells developed independently from PD-1⁺ cells and not in direct contact with B cells. More importantly, CD62L⁺ Tfh cells expressed memory- and stemness-associated genes, and with better superior long-term survival, they readily generated PD-1hi cells in the recall response. Finally, KLF2 and IL7R, also highly expressed by CD62L⁺ Tfh cells, were required to regulate their development. Our work thus demonstrates a novel Tfh memory-like cell subpopulation, which may benefit our understanding of immune responses and diseases.
... To recall immune responses against previously encountered pathogens, memory cells need to be maintained even in the absence of antigenic exposure [79]. Homeostatic proliferation and survival of T cells are driven by cytokines such as IL-7 and IL-15 [80][81][82][83][84]. Indeed, these cytokines also play a role in the persistence of infected CD4 + T cells. ...
Article
Full-text available
Purpose of Review In this review, we discuss what persistent viremia has taught us about the biology of the HIV-1 reservoir during antiretroviral therapy (ART). We will also discuss the implications of this phenomenon for HIV-1 cure research and its clinical management. Recent Findings While residual viremia (RV, 1–3 HIV-1 RNA copies/ml) can be detected in most of people on ART, some individuals experience non-suppressible viremia (NSV, > 20–50 copies/mL) despite optimal adherence. When issues of drug resistance and pharmacokinetics are ruled out, this persistent virus in plasma is the reflection of virus production from clonally expanded CD4⁺ T cells carrying proviruses. Recent work has shown that a fraction of the proviruses source of NSV are not infectious, due to defects in the 5′-Leader sequence. However, additional viruses and host determinants of NSV are not fully understood. Summary The study of NSV is of prime importance because it represents a challenge for the clinical care of people on ART, and it sheds light on virus-host interactions that could advance HIV-1 remission research.
... 18-20 IL-7/IL-7 receptor signaling, which increases expression of anti-apoptotic proteins that include Bcl-2, is particularly important for the generation of long-lived virus-specific CD4 + and CD8 + T cells in humans and animal models. 7,19,[21][22][23][24] The IL-7 receptor is comprised of the common g chain and an a chain, CD127. In HIV infection, longlived T cells expressing CD127 are lost, 19,25,26 contributing to loss of immunological memory, evidenced by poor responses to vaccination. ...
Article
Full-text available
In people with HIV (PWH), the post-antiretroviral therapy (ART) window is critical for immune restoration and HIV reservoir stabilization. We employ deep immune profiling and T cell receptor (TCR) sequencing and examine proliferation to assess how ART impacts T cell homeostasis. In PWH on long-term ART, lymphocyte frequencies and phenotypes are mostly stable. By contrast, broad phenotypic changes in natural killer (NK) cells, γδ T cells, B cells, and CD4⁺ and CD8⁺ T cells are observed in the post-ART window. Whereas CD8⁺ T cells mostly restore, memory CD4⁺ T subsets and cytolytic NK cells show incomplete restoration 1.4 years post ART. Surprisingly, the hierarchies and frequencies of dominant CD4 TCR clonotypes (0.1%–11% of all CD4⁺ T cells) remain stable post ART, suggesting that clonal homeostasis can be independent of homeostatic processes regulating CD4⁺ T cell absolute number, phenotypes, and function. The slow restoration of host immunity post ART also has implications for the design of ART interruption studies.
... High levels of CD44, IL-7R, and IL-15R are recognized as both phenotypic and functional markers for memory T cells. 15,[27][28][29][30] In addition, both CD44 hi and CD44 -~lo CD4 + T-cell populations were present in the relevant SLO of CAU mice (ELN, ILN, and spleen), and CD44 hi CD4 + T cells similarly exhibited co-expressions of IL-7R and IL-15R, in contrast to their CD44 -~lo CD4 + counterpart ( Figure 1C), suggesting that uveitis-associated memory CD4 + T cells also reside in these lymphoid tissues in chronic uveitis. ...
Article
Full-text available
Chronic uveitis comprises heterogeneous clinical entities characterized by sustained and recurrent intraocular inflammation that is believed to be driven by autoimmune responses. The management of chronic uveitis is challenging with the limited availability of efficacious treatments, and the underlying mechanisms mediating disease chronicity remain poorly understood as the majority of experimental data are derived from the acute phase of the disease (the first 2–3 weeks post‐induction). Herein, we investigated the key cellular mechanisms underlying chronic intraocular inflammation using our recently established murine model of chronic autoimmune uveitis. We demonstrate unique long‐lived CD44hiIL‐7R⁺IL‐15R⁺CD4⁺ memory T cells in both retina and secondary lymphoid organs after 3 months postinduction of autoimmune uveitis. These memory T cells functionally exhibit antigen‐specific proliferation and activation in response to retinal peptide stimulation in vitro. Critically, these effector‐memory T cells are capable of effectively trafficking to the retina and accumulating in the local tissues secreting both IL‐17 and IFN‐γ upon adoptively transferred, leading to retinal structural and functional damage. Thus, our data reveal the critical uveitogenic functions of memory CD4⁺ T cells in sustaining chronic intraocular inflammation, suggesting that memory T cells can be a novel and promising therapeutic target for treating chronic uveitis in future translational studies.
... In fact, the raising of T and B cells and the rapid increase of IL-7Ra-expressing T cells indicated the development and the maintenance of memory pools (17,18) and anticipated the immune features found in the LL. ...
Article
Full-text available
The current level of evidence for immunotherapy in previously untreated microsatellite unstable metastatic colorectal cancer is based on recent pieces of evidence of few studies that demonstrated durable response and clinical benefit, in terms of objective response rate, disease control rate, and progression-free survival in this subgroup of patients. On the basis of combinatorial immunotherapy with nivolumab plus ipilimumab, we report the exceptional case of a complete pathological response in a 21-year-old woman presenting a clinically aggressive stage IV colorectal cancer with massive nodal and liver involvement. Extensive molecular analyses based on whole genome next-generation DNA sequencing, RNA sequencing, fluorescent multiplex immunohistochemistry, and flow cytometry provided a detailed description of tumoral and immunological characteristics of this noteworthy clinical case.
Article
Full-text available
Successful antiretroviral therapy (ART) can efficiently suppress Human Immunodeficiency Virus-1 (HIV-1) replication to undetectable levels, but rare populations of infected memory CD4 ⁺ T cells continue to persist, complicating viral eradication efforts. Memory T cells utilize distinct homing and adhesion molecules to enter, exit, or establish residence at diverse tissue sites, integrating cellular and environmental cues that maintain homeostasis and life-long protection against pathogens. Critical roles for T cell receptor and cytokine signals driving clonal expansion and memory generation during immunity generation are well established, but whether HIV-infected T cells can utilize similar mechanisms for their own long-term survival is unclear. How infected, but transcriptionally silent T cells maintain their recirculation potential through blood and peripheral tissues, or whether they acquire new capabilities to establish unique peripheral tissue niches, is also not well understood. In this review, we will discuss the cellular and molecular cues that are important for memory T cell homeostasis and highlight opportunities for HIV to hijack normal immunological processes to establish long-term viral persistence.
Article
Full-text available
Kidney transplantation is a well-established alternative to renal replacement therapy. Although the number of patients with end-stage renal disease (ESRD) is increasing, the availability of kidney for transplantation is still insufficient to meet the needs. As age increases, the prevalence of ESRD increases; thus, the population of aged donors and recipients occupies large proportion. Accumulated senescent cells secrete pro-inflammatory factors and induce senescence. Additionally, it is gradually becoming clear that biological sex differences can influence aging and cause differences in senescence. Here, we review whether age-related sex differences affect organ transplant outcomes and what should be done in the future.
Chapter
Here we review the contributions of several members of the tumor necrosis factor receptor super family (TNFRSF) to CD4 T cell responses, focusing on those that govern the survival of responding cells and the establishment of a functional memory pool.
Article
Overview IL-7 is a member of the family of cytokines with four anti-parallel α helixes that bind Type I cytokine receptors. It is produced by stromal cells and is required for development and homeostatic survival of lymphoid cells. Genomic architecture Interleukin 7 (IL7) human IL7: gene ID: 3574 on ch 8; murine Il7 gene ID: 16,196 on ch 3. Protein Precursor contains a signal sequence, mature human IL-7 peptide 152aa, predicted 17.4kd peptide, glycosylated resulting in 25kd. Crystal structure: http://www.rcsb.org/structure/3DI2. Regulation of IL-7 production Major producers are stromal cells in thymus, bone marrow and lymphoid organs but also reported in other tissues. Production is primarily constitutive but reported to be affected by IFNγ and other factors. IL-7 receptors Two chains IL-7Rα (IL-7R) and γc (IL-2RG). Human IL-7R: gene ID 3575 on ch 5; human IL2RG: gene ID 3561 on ch X; mouse IL-7R: gene ID 16,197 on ch 15; murine Il2rg gene ID 16,186 on ch X. Member of γc family of receptors for cytokines IL-2, −4, −9, −15, and −21. Primarily expressed on lymphocytes but reports of other cell types. Expression in T-cells downregulated by IL-7. Low expression on Tregs, no expression on mature B-cells. Crystal structure: http://www.rcsb.org/structure/3DI2. IL-7 receptor signal transduction pathways Major signals through JAK1, JAK3 to STAT5 and through non-canonical STAT3, STAT1, PI3K/AKT and MEK/ERK pathways. Biological activity of IL-7 Required for survival of immature thymocytes, naïve T-cells, memory T-cells, pro-B-cells and innate lymphocytes. Pharmacological treatment with IL-7 induces expansion of naïve and memory T-cells and pro-B-cells. Abnormalities of the IL-7 pathway in disease Deficiencies in the IL-7 pathway in humans and mice result in severe combined immunodeficiency due to lymphopenia. Excessive signaling of the pathway in mice drives autoimmune diseases and in humans is associated with autoimmune syndromes including multiple sclerosis, type 1 diabetes, rheumatoid arthritis, sarcoidosis, atopic dermatitis and asthma. Mutations in the IL-7 receptor pathway drive acute lymphoblastic leukemia. Clinical applications IL-7 has been evaluated in patients with cancer and shown to expand lymphocytes. It accelerated lymphocyte recovery after hematopoietic stem cell transfer, and increased lymphocyte counts in AIDS patients and sepsis patients. Monoclonal antibodies blocking the IL-7 receptor are being evaluated in autoimmune diseases. Cytotoxic monoclonals are being evaluated in acute lymphoblastic leukemia. Drugs blocking the signal transduction pathway are being tested in autoimmunity and acute lymphoblastic leukemia.
Article
Full-text available
In the immune system, there is a careful regulation not only of lymphoid development and proliferation, but also of the fate of activated and proliferating cells. Although the manner in which these diverse events are coordinated is incompletely understood, cytokines are known to play major roles. Whereas IL-7 is essential for lymphoid development, IL-2 and IL-4 are vital for lymphocyte proliferation. The receptors for each of these cytokines contain the common cytokine receptor γ chain (γc), and it was previously shown that γc-deficient mice exhibit severely compromised development and responsiveness to IL-2, IL-4, and IL-7. Nevertheless, these mice exhibit an age-dependent accumulation of splenic CD4+ T cells, the majority of which have a phenotype typical of memory/activated cells. When γc-deficient mice were mated to DO11.10 T cell receptor (TCR) transgenic mice, only the T cells bearing endogenous TCRs had this phenotype, suggesting that its acquisition was TCR dependent. Not only do the CD4+ T cells from γc-deficient mice exhibit an activated phenotype and greatly enhanced incorporation of bromodeoxyuridine but, consistent with the lack of γc-dependent survival signals, they also exhibit an augmented rate of apoptosis. However, because the CD4+ T cells accumulate, it is clear that the rate of proliferation exceeds the rate of cell death. Thus, surprisingly, although γc-independent signals are sufficient to mediate expansion of CD4+ T cells in these mice, γc-dependent signals are required to regulate the fate of activated CD4+ T cells, underscoring the importance of γc-dependent signals in controlling lymphoid homeostasis.
Article
Full-text available
Interleukin (IL)-7 is a potent stimulus for immature T and B cells and, to a lesser extent, mature T cells. We have inactivated the IL-7 gene in the mouse germline by using gene-targeting techniques to further understand the biology of IL-7. Mutant mice were highly lymphopenic in the peripheral blood and lymphoid organs. Bone marrow B lymphopoiesis was blocked at the transition from pro-B to pre-B cells. Thymic cellularity was reduced 20-fold, but retained normal distribution of CD4 and CD8. Splenic T cellularity was reduced 10-fold. Splenic B cells, also reduced in number, showed an abnormal population of immature B cells in adult animals. The remaining splenic populations of lymphocytes showed normal responsiveness to mitogenic stimuli. These data show that proper T and B cell development is dependent on IL-7. The IL-7-deficient mice are the first example of single cytokine-deficient mice that exhibit severe lymphoid abnormalities.
Article
Interleukin (IL)-7 is a potent stimulus for immature T and B cells and, to a lesser extent, mature T cells. We have inactivated the IL-7 gene in the mouse germline by using gene-targeting techniques to further understand the biology of IL-7. Mutant mice were highly lymphopenic in the peripheral blood and lymphoid organs. Bone marrow B lymphopoiesis was blocked at the transition from pro-B to pre-B cells. Thymic cellularity was reduced 20-fold, but retained normal distribution of CD4 and CD8. Splenic T cellularity was reduced 10-fold. Splenic B cells, also reduced in number, showed an abnormal population of immature B cells in adult animals. The remaining splenic populations of lymphocytes showed normal responsiveness to mitogenic stimuli. These data show that proper T and B cell development is dependent on IL-7. The IL-7-deficient mice are the first example of single cytokine-deficient mice that exhibit severe lymphoid abnormalities.
Article
The electronic structures of various Cu+ luminescence centers in wurtzite ZnS are studied by using first-principles linear muffin-tin-orbital method combining atomic sphere approximation. The results for ZnS:Cu:Cl and ZnS:Cu:Al show that Cu acceptor states are anomalously deep (beyond 2.7 eV above the top of valence band) and located near the bottom of conduction band in these two cases. On the other hand, the results for ZnS:Cu with sulfur vacancies manifest that the Cu+ center is an associated center, whose Cu d-like states are situated above the top of valence band. Bibtex entry for this abstract Preferred format for this abstract (see Preferences) Find Similar Abstracts: Use: Authors Title Abstract Text Return: Query Results Return items starting with number Query Form Database: Astronomy Physics arXiv e-prints
Article
The requisite molecular interactions for CD8 T cell memory were determined by comparison of monoclonal naı̈ve and memory CD8+ T cells bearing the T cell receptor (TCR) for the HY antigen. Naı̈ve T cells required only the right major histocompatibility complex (MHC) class I–restricting molecule to survive; to expand, they also needed antigen. In contrast, for survival, memory cells did not require the restricting MHC allele, but needed only a nonspecific class I; for expansion the correct class I, but not antigen, was required. Thus, maintenance of CD8 T cell memory still required TCR–MHC class I interactions, but memory T cells may have a lower functional activation threshold that facilitates secondary responses.
Article
Biochemical studies of signaling mediated by many cytokine and growth factor receptors have implicated members of the Jak family of tyrosine kinases in these pathways. Specifically, Jak3 has been shown to be associated with the interleukin-2 (IL-2) receptor γ chain, a component of the receptors for IL-2, IL-4, IL-7, IL-9, and IL-15. Mice lacking Jak3 showed a severe block in B cell development at the pre-B stage in the bone marrow. In contrast, although the thymuses of these mice were small, T cell maturation progressed relatively normally. In response to mitogenic signals, peripheral T cells in Jak3-deficient mice did not proliferate and secreted small amounts of IL-2. These data demonstrate that Jak3 is critical for the progression of B cell development in the bone marrow and for the functional competence of mature T cells.
Article
The immune system relies on homeostatic mechanisms in order to adapt to the changing requirements encountered during steady-state existence and activation by antigen. For T cells, this involves maintenance of a diverse repertoire of naïve cells, rapid elimination of effector cells after pathogen clearance, and long-term survival of memory cells. The reduction of T-cell counts by either cytotoxic drugs, irradiation, or certain viruses is known to lead to lymphopenia-induced proliferation and restoration of normal T-cell levels. Such expansion is governed by the interaction of TCR with self-peptide/MHC (p/MHC) molecules plus contact with cytokines, especially IL-7. These same ligands, i.e. p/MHC molecules and IL-7, maintain naïve T lymphocytes as resting cells under steady-state T-cell-sufficient conditions. Unlike naïve cells, typical "central" memory T cells rely on a combination of IL-7 and IL-15 for their survival in interphase and for occasional cell division without requiring signals from p/MHC molecules. Other memory T-cell subsets are less quiescent and include naturally occurring activated memory-phenotype cells, memory cells generated during chronic viral infections, and effector memory cells. These subsets of activated memory cells differ from central memory T cells in their requirements for homeostatic proliferation and survival. Thus, the factors controlling T-cell homeostasis can be seen to vary considerably from one subset to another as described in detail in this review.
Article
We have concentrated here on the lymphokines which might serve to regulate the different pathways of precursor development. We suggest that, as a result of antigenic stimulation, specific precursor cells both proliferate and become committed to develop into either an effector cell, a memory cell or an anergized cell. Anergy has not been dealt with in this review, but it is likely to be one of the options available. The development of an effector population takes 4-7 d (quite analogous to the time it takes for CTLp to become CTL and for resting B to become Ab-forming cells). The effector populations are large, generally IL-2R-positive cells. These cells have upregulated many adhesion molecule systems [e.g., Pgp-1, LFA-1 and ICAM-1 (Swain unpublished)], but downregulated the Mel-14 homing receptor. Effectors are ready to respond to APC such as specific B cells with a rapid synthesis and secretion of lymphokines. The effector population is then quickly downregulated, both by the turn off of lymphokine synthesis/secretion and possibly by its own suicide. This kind of pattern makes teleological sense since the cells making such high titers of lymphokines could have many potent pleitropic effects. It also seems to be the strategy employed in the generation of other terminally differentiated effectors (such as CTL and plasma cells). The requirement for restimulation and the requirement for direct and perhaps prolonged contact between the helper effector and the APC-B cell can be expected to help ensure that these lymphokines are localized (reviewed in Swain & Dutton 1987, Swain & Croft 1990) and effectively delivered to specific responding cells. We postulate that at the same time, or perhaps subsequent to this, another set of signals drives precursors to generate prememory cells. Our studies suggest these emerging memory cells may be phenotypically unique and we postulate that they are specialized to become a "long-lived" population of memory cells that will persist indefinitely as a protective population of increased frequency for the antigen encountered and which is also able to respond more rapidly and effectively. The greater effectiveness of the memory response would thus be due to dramatically increased frequency, to characteristic and stable changes in adhesion molecule expression and to the fact that, in addition to IL-2, resting memory cells also secrete at least low titers of IL-3, IL-4, IFN-gamma and other lymphokines upon initial restimulation.(ABSTRACT TRUNCATED AT 400 WORDS)
Article
The common gamma chain (gamma c) of the IL-2, IL-4, IL-7, IL-9, and IL-15 receptors is defective in humans with XSCID. Mice lacking gamma c expression had hypoplastic thymuses; the thymocytes responded to gamma c-independent mitogens, but not gamma c-dependent stimuli. Splenic T cells were diminished at 3 weeks of age, but CD4+ T cells markedly increased by 4 weeks. B cells were greatly diminished in contrast with the situation in XSCID. NK cells, gamma delta intestinal intraepithelial lymphocytes, dendritic epidermal T cells, peripheral lymph nodes, and gut-associated lymphoid tissue were absent. These findings underscore the importance of gamma c in lymphoid development. Moreover, differences in humans and mice lacking gamma c expression indicate species-specific differences in the roles of gamma c-dependent cytokines or in the existence of redundant pathways. These mice provide an important model for studying the pathophysiology provide an important model for studying the pathophysiology of and gene therapy for human XSCID.