ArticlePDF Available

Effects of Increasing IL-7 Availability on Lymphocytes during and after Lymphopenia-Induced Proliferation

Authors:
  • Inserm (National Institute of Health and Medical Research)

Abstract and Figures

IL-7 is critically involved in regulating peripheral T cell homeostasis. To investigate the role of IL-7 on lymphopenia-induced proliferation of polyclonal lymphocytes, we have transferred CFSE-labeled cells into a novel T-lymphopenic, IL-7-transgenic mouse line. Results obtained indicate that T and B cells do not respond in the same way to IL-7-homeostatic signals. Overexpression of IL-7 enhances proliferation of both CD4(+) and CD8(+) T cells but with distinctly temporal effects. Expansion of naturally arising CD4(+)-regulatory T cells was like that of conventional CD4(+) T cells. IL-7 had no effect on B cell proliferation. By immunohistology, transferred T cells homed to T cell areas of spleen lymphoid follicles. Increasing IL-7 availability enhanced T cell recovery by promoting cell proliferation and reducing apoptosis during early stages of lymphopenia-induced proliferation. Taken together, these results provide new insights into the pleiotropic effects of IL-7 on lymphopenia-induced T cell proliferation.
Content may be subject to copyright.
Effects of Increasing IL-7 Availability on Lymphocytes during
and after Lymphopenia-Induced Proliferation
1
Nabil Bosco,* Fabien Agene`s,* and Rhodri Ceredig
2†
IL-7 is critically involved in regulating peripheral T cell homeostasis. To investigate the role of IL-7 on lymphopenia-induced
proliferation of polyclonal lymphocytes, we have transferred CFSE-labeled cells into a novel T-lymphopenic, IL-7-transgenic
mouse line. Results obtained indicate that T and B cells do not respond in the same way to IL-7-homeostatic signals. Overex-
pression of IL-7 enhances proliferation of both CD4
and CD8
T cells but with distinctly temporal effects. Expansion of naturally
arising CD4
-regulatory T cells was like that of conventional CD4
T cells. IL-7 had no effect on B cell proliferation. By
immunohistology, transferred T cells homed to T cell areas of spleen lymphoid follicles. Increasing IL-7 availability enhanced T
cell recovery by promoting cell proliferation and reducing apoptosis during early stages of lymphopenia-induced proliferation.
Taken together, these results provide new insights into the pleiotropic effects of IL-7 on lymphopenia-induced T cell
proliferation. The Journal of Immunology, 2005, 175: 162–170.
Despite declining thymic output with age, the peripheral T
cell pool of an adult animal remains remarkably stable
(1, 2). How the T cell pool is maintained remains a cen-
tral question in immunology. Compelling data have been provided
indicating that long term survival and homeostatic proliferation of
T lymphocytes is dependent on a combination of low level TCR
and cytokine stimulation. After transfer into a lymphopenic envi-
ronment, T cells sense the absence of T cells and proliferate
slowly, a process that has been termed lymphopenia-induced pro-
liferation (LIP)
3
(3). Cytokines, such as IL-7 and IL-15, have been
shown to play a major role in both LIP and T cell survival in mice
(1, 4). IL-7 is also a crucial cytokine for lymphocyte development
providing survival-promoting signals for immature and mature T
as well as for immature B cells (5).
Different experimental procedures have been designed to study
the role of IL-7 in LIP. IL-7 enhances survival of mature T cells in
vitro (6, 7), and exogenous IL-7 administration increases both the
pool size of peripheral T cells (8) and the rate of hemopoietic
reconstitution after bone marrow (BM) transplantation in vivo (9).
Nevertheless, when IL-7 is injected into normal mice, it has been
difficult to distinguish between its effects on thymic output and that
on peripheral T cells. Syngenic adoptive transfer experiments into
either IL-7-deficient or wild-type mice treated with anti-IL-7 or
anti-IL-7R
mAb show that perturbation of IL-7 signals prevents
the expansion of transferred T cells (1, 10). Although several
groups have shown that IL-7 is crucial for T cell survival and LIP,
the mechanisms by which IL-7 levels regulate the size and diver-
sity of the peripheral T cell pool are still not well understood.
Differences in experimental systems used to investigate the role
of IL-7 in LIP may account for some apparently conflicting data.
These differences concern either the nature of the recipient mouse
or that of the transferred cells. Preconditioning the recipient mouse
by irradiation or other lymphocyte-depleting regimens probably
alters lymphoid organ architecture, Ag-presenting cell function,
and/or cytokine milieu (11, 12). For instance, after irradiation, IL-7
and TGF
levels may change, thereby affecting LIP (12, 13). Fur-
thermore, in sublethally irradiated recipients, residual bystander T
cells persist and compete with transferred T cells for cytokine and
or self peptide-MHC complexes, thereby influencing reconstitu-
tion of the T cell compartment (12). Transferred cells are fre-
quently derived from TCR-transgenic, recombinase-activating
gene-deficient (RAG
/
) mice. Such monoclonal T cell popula-
tions express a TCR of predefined specificity and affinity, two
parameters that correlate with CD5 expression and that may dictate
whether a T cell undergoes LIP (14 –16). Second, such monoclonal
T cell populations lack naturally arising CD4
CD25
-regulatory
T cells (17, 18) known to alter the behavior of cells during LIP (19,
20). In such experiments, long term survival is rarely monitored
because for various reasons, including the absence of
CD4
CD25
-regulatory T cells, recipients frequently develop au-
toimmune diseases (21, 22).
Under normal T cell-replete, nonlymphopenic conditions, IL-7
probably acts as a survival factor for T cells (4). In normal mice,
the net level of IL-7 availability is low, resulting both from limited
production by stromal cells and simultaneous consumption by T
cells (23). In T cell-lymphopenic conditions, the net IL-7 level
increases by a poorly defined mechanism, thereby allowing resid-
ual T cells to sense lymphopenia, augment TCR signaling, and
consequently triggering LIP. This notion is consistent with avail-
able data and particularly with the previously reported phenotype
of IL-7-transgenic (IL-7Tg) mice (24) that contain a stable ex-
panded (20-fold) T cell pool (25, 26). It could be postulated that
net IL-7 availability provides the main clue whereby T cells sense
*Institut National de la Sante´ et de la Recherche Me´dicale Unite´ 548, De´partement de
Re´ponse et Dynamique Cellulaire, Commissariat a` l’Energie Atomique-G, Grenoble,
France; and
Institut National de la Sante´ et de la Recherche Me´dicale Unite´ 645,
Institut Federatif de Recherche 133, Etablissement Franc¸ais du Sang, Besanc¸on,
France
Received for publication January 4, 2005. Accepted for publication April 21, 2005.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
1
This work was supported by institutional grants from Institut National de la Sante´
et de la Recherche Me´dicale and the Commissariat a` l’Energie Atomique, a specific
grant “The´matiques Prioritaires de la Re´gion Rhoˆne-Alpes.” M.N.B. has a PhD schol-
arship from the Commissariat a` l’Energie Atomique.
2
Address correspondence and reprint requests to Dr. Rod Ceredig, Institut National
de la Sante´ et de la Recherche Me´dicale Unite´ 645, Institut Federatif de Recherche
133, Etablissement Franc¸ais du Sang, 1 Boulevard Alexander Fleming, 25020 Be-
sanc¸on, France. E-mail address: Rod.Ceredig@efs.sante.fr
3
Abbreviations used in this paper: LIP, lymphopenia-induced proliferation; LN,
lymph nodes; PALS, periarteriolar lymphocyte sheaths; RAG-2, recombinase-acti-
vating gene-2; Tg, transgenic; SP, CD4
or CD8
single-positive; BM, bone marrow;
CEA, Commissariat a` l’Energie Atomique; mEF1
, mouse elongation factor 1
.
The Journal of Immunology
Copyright © 2005 by The American Association of Immunologists, Inc. 0022-1767/05/$02.00
lymphopenia. Thus, in IL-7Tg mice, T cells increase in number
until capable of absorbing the increased available IL-7. Recently,
Li et al. (27) predicted that in lymphopenic recipients, where IL-7
availability was increased, T cell LIP would be triggered. How-
ever, to date, this hypothesis has not been directly demonstrated,
and no study has systematically examined the behavior of poly-
clonal T and B cells transferred together into mice differing in IL-7
availability.
For the studies reported herein, we have developed a novel T
cell lymphopenic mouse strain that overexpresses IL-7. The IL-7
transgene was introduced into C57BL/6.CD3
gene-deficient
(CD3
/
) mice (28). IL-7Tg.CD3
/
and nontransgenic
CD3
/
littermate controls provide a pair of T-lymphopenic, B
lymphocyte-containing mice differing only in net IL-7 availability
and not requiring further conditioning before use as recipients. To
investigate how IL-7 availability influences the behavior of cells
undergoing LIP in T-lymphopenic animals, we transferred CFSE-
labeled polyclonal T and B cells into recipient mice. Parameters
investigated included the kinetics of both CD4
and CD8
T cell
as well as B cell growth and division, anatomical localization, the
kinetics of Bcl-2 expression and cell loss by apoptosis, as well as
long term cell recovery and turnover. Taken together, our results
provide new insights into the dynamics of IL-7-dependent LIP and
the pleiotropic effects of IL-7 on T and B cell homeostasis.
Materials and Methods
Mice
IL-7-transgenic mice on a C57BL/6 (B6) genetic background (IL-7Tg.B6)
have been described previously (24); they were maintained in a heterozy-
gous state by breeding transgenic males with B6 females (Iffa-Credo). IL-
7Tg.CD3
/
(28) and IL-7Tg.RAG-2
/
mice (29) were generated in the
animal facility of the De´partement de Re´ponse et Dynamique Cellulaires,
Commissariat a` l’Energie Atomique (CEA)-Grenoble by intercrossing IL-
7Tg.B6 males with the respective knockout females, obtained from CDTA
France. Mice were screened for the IL-7 transgene by PCR on tail DNA
and FACS of PBL. Once obtained, IL-7Tg.CD3
/
or IL-7Tg.RAG-2
/
mice were maintained by intercrossing the respective IL-7Tg males with
nontransgenic knockout females. P14.RAG-2
/
mice (30) were a gift of
Dr. Jo¨rg Kirberg (Max Planck Institute for Immunobiology, Freiburg, Ger-
many). In most experiments, 6- to 8-wk-old mice were used and were
heterozygous for the IL-7 transgene. Animal care and experimental pro-
cedures conformed to those of the CEA-Grenoble animal care and users
committee.
Flow cytometry
The following mAbs, obtained from BD Pharmingen or e-Bioscience, were
used for staining: anti-CD4 (GK1.5); anti-CD8 (53-6.7); anti-CD19 (1D3);
anti-CD21 (7G6); anti-CD23 (B3B4); anti-CD45R (RA3-6B2); anti-CD25
(7D4); anti-CD44 (IM7); anti-CD62L (MEL-14); anti-CD69 (H1-2F3); anti-
CD117 or c-Kit (2B8); anti-CD122 (TM-
1); anti-CD127 (A7R34); anti-
CD132 (4G3); anti-NK1.1 (PK136); and anti-IgM (R6-60.2). Cells were
three- or four-color stained with appropriate combinations of FITC-, PE-,
Cy-, and biotin-labeled Abs, followed by streptavidin-APC (BD Pharmin-
gen). Dead cells were excluded from analysis by light scatter and when
possible propidium iodide staining. All analyses were performed using a
FACSCalibur flow cytometer and data analyzed using CellQuest (BD Bio-
sciences) or WinMDI (Joseph Trotter) software.
Adoptive transfer experiments
Single-cell suspensions of lymph nodes (LN) and spleen lymphocytes were
prepared and purified over a Ficoll gradient before counting viable cells by
trypan blue exclusion and labeling with 5
M CFSE (Molecular Probes) as
described previously (31). Labeled lymphocytes (10
7
) were administered
i.v. to unirradiated lymphopenic mice. Mice were sacrificed at different
indicated times after adoptive transfer. Then, spleen, LN, and thymus were
collected, and single-cell suspensions were prepared and counted before
being surface stained as described above. CFSE and surface staining were
then analyzed by FACS. The total number of each T cell subpopulation was
calculated as described above from the frequency determined by FACS,
and the total number of viable cells recovered in each organ determined by
trypan blue exclusion. For naive T cell transfer experiments, naive
CD4
CD45RB
high
and CD8
CD44
low
T cells were sorted from spleen of
8-wk-old B6 mice with a Moflo (Cytomation) with a purity of 97%, then
labeled with CFSE, and cotransferred as described above.
Apoptosis assay
Externalization of phosphatidylserine was detected by PE-conjugated an-
nexin V mAb using the apoptosis detection kit (BD Pharmingen) according
to the instructions of the manufacturer. In brief, 10
6
splenocytes from adop-
tively transferred mice at day 3 or 28 were surface stained and then washed
with binding buffer. Cells were incubated for 15 min at room temperature
in the dark with annexin V 7-aminoactinomycin D in binding buffer.
Then, 10
6
cells were analyzed by FACS as described above.
In vivo BrdU labeling
At 23 days after T cell transfer, mice were given 1 mg of BrdU (Sigma-
Aldrich) by i.p. injection and 1 mg/ml BrdU in their drinking water for 5
days. Recipient mice were sacrificed at day 28 after T cell transfer, and
spleen cell suspensions were prepared, surface stained as described above,
washed, fixed, and permeabilized before labeling with anti-BrdU mAb as
described previously (32, 33) using the BrdU flow kit (BD Pharmingen).
Then, 10
6
cells were analyzed by FACS.
Intracellular staining for Bcl-2
As described above, 10
6
total spleen cells from unmanipulated B6 or adop-
tively transferred mice were prepared and surface stained. After unbound
mAb were washed, cells were subjected to intracellular staining for Bcl-2
using the Cytofix/Cytoperm kit (BD Pharmingen). For intracellular stain-
ing, FITC-conjugated hamster anti-mouse Bcl-2 mAb (clone 3F11) or an
isotype control mAb (hamster IgG) was used.
Quantitative RT- PCR for IL-7 expression
Quantification of IL-7 transcript was performed on 6- to 8-wk-old mice.
RNA was extracted from 2 to 5 10
6
thymus or spleen cells of IL-7
transgenic mice or their negative littermate controls. RNA was extracted
(Trizol; Invitrogen), DNase digested (DNase I; Invitrogen) and cDNA syn-
thesized using SuperScriptII reverse transcriptase (Invitrogen). Real time
quantitative PCR was performed using a Lightcycler Instrument using the
FastStart kit, 1U/reaction (Roche Diagnostics). The real time PCR condi-
tions were as follows: 95°C for 8 min; then 45 cycles with 95°C for 15 s
at 60°C for 1 min. Mouse elongation factor 1
(mEF1
) mRNA was used
as a positive control and to quantitate total amplified RNA. Analysis was
conducted with Light Cycler 3.5 software (Roche Diagnostics). Results
were expressed as the ratio of IL-7 mRNA to mEF1
mRNA and arbi-
trarily set at 1 for reference mice. The primer (Eurogentec) sequences were
as follows: mIL7 sense, 5-CAGACCATGTTCCATGTTTCTTTTA-3;
mIL7 antisense, 5-CTTTGTCTTTAATGTGGCACTCAGA-3; mEF1
sense, 5-CTGAACCATCCAGGCCAAAT-3; and mEF1
antisense,
5-TCTTTTCTTTAAGCTCAGCAAACTTG-3.
Immunohistochemistry
Spleens were harvested from recipient mice at different times after adoptive
transfer, embedded in Cryo-M-Bed compound (Bright Instrument), and
frozen at 80°C. Frozen sections 5–7
m thick were fixed for 10 min in
cold acetone and dried extensively. Sections were stained with PE-labeled
anti-B220 mAb (RA3-6B2) for B cells and, when CFSE staining had be-
come negative, FITC-labeled anti-CD90 mAb (T24) to detect T cells. Cells
were imaged using a fluorescent confocal microscope (Leica TCS-SP3).
Results
Characterization of lymphopenic recipient mice
To study the effect of increased IL-7 availability on LIP, two models
of lymphopenic mice overexpressing IL-7 were generated. For most
of the experiments reported herein, we used only IL-7Tg.CD3
/
and CD3
/
littermates. The expression of the mouse IL-7 cDNA
transgene is controlled by the mouse MHC class II promoter (24), and
MHC class II genes are actively transcribed in mouse B cells (34).
Because B cells are present in CD3
/
mice, we observed by quan-
titative RT-PCR that differences in IL-7 transcript levels were greater
in spleen and thymus between IL-7Tg.CD3
/
and CD3
/
litter-
mates (10- to 30-fold) than between IL-7Tg.RAG-2
/
and RAG-
2
/
littermates (3- to 5-fold) where B cells are absent (not shown).
163The Journal of Immunology
Overexpression of IL-7 perturbs B cell development, primarily
by expanding the progenitor pool in the BM (35, 36). To demon-
strate this in the IL-7Tg.CD3
/
mouse line and because no re-
liable assay exists to quantitate available IL-7 protein in organs, a
series of phenotypic analyses was performed. The total number of
CD19
B cells was increased 6-fold in the BM of IL-
7Tg.CD3
/
mice compared with littermate controls (Fig. 1A).
Both CD117
pro-B/pre-BI and CD25
B220
IgM
pre-BII cells
were increased 10-fold; the numbers of B220
IgM
low
immature
B and IgM
⫹⫹
or CD23
mature B cells remained unchanged.
Thus, the known IL-7-dependent stages of BM B cell development
were the most affected by MHC class II promoter-driven IL-7
overexpression (32, 35, 36).
Two major changes were noted in the spleens of adult IL-
7Tg.CD3
/
mice. First, the spleen contained large numbers of
IL-7-dependent CD117
pro/pre-BI cells (Fig. 1B, bar graph) and
B220
IgM
immature B cells (Fig. 1B, cytograms); such cells are
only normally found in neonatal mice (37). Second, although
CD23
CD21
follicular B cell numbers were similar, there were
fewer CD23
low
CD21
marginal zone B cells in IL-7Tg.CD3
/
mice (Fig. 1B, lower cytograms and bar graph). Importantly, con-
focal microscopy indicated that spleen architecture was largely
preserved (see below). Qualitatively, Ab responses to the T-inde-
pendent type II Ag trinitrophenyl-Ficoll were normal in IL-
7Tg.CD3
/
and CD3
/
littermate controls, but IgM and IgG
titers were 10-fold higher in transgenics (Ref. 32 and data not
shown). In the peritoneal cavity, CD23
CD5
B2 B cells were
increased 4-fold whereas in the peripheral blood, B220
IgM
pre-
BII and B220
IgM
low
immature cells were present (Ref. 32 and
data not shown).
Analysis of thymocytes as reviewed by Ceredig and Rolink
(33) indicated that the numbers of IL-7-responsive
CD117
⫹⫹
CD44
CD25
DN1 and CD44
, CD25
DN2 cells
were increased 2-fold but that the number of CD44
CD25
DN3 thymocytes remained unchanged (Fig. 1C) (38). Signifi-
cantly, the proportion of total CD44
CD25
DN1 cells was in-
creased with many cells staining weakly for CD44 (Fig. 1C, upper
cytograms). As expected (37), additional analysis indicated that
these CD44
low
CD25
cells were CD19
B cells comprising a
mixture of B220
IgM
pre-BII and B220
IgM
low
immature B
cells. CD19
B cells in the thymus of CD3
/
mice were mature
IgM
cells (Fig. 1C, lower cytograms). Taken together, these phe-
notypic and quantitative PCR data indicate that in IL-
7Tg.CD3
/
mice, BM B cell development was drastically al-
tered, and immature B cells were present in the blood, spleen, and
even the thymus. These changes reflect the increased IL-7 avail-
ability in these mice.
Increase of IL-7 availability promotes polyclonal T cell recovery
in vivo
To carefully characterize the contribution of IL-7 to T cell ho-
meostasis, unirradiated IL-7Tg.CD3
/
and their CD3
/
lit-
termates received i.v. 10
7
pooled LN and spleen lymphocytes, con-
taining 2–3 10
6
CD4
and CD8
T cells. Recipients were
sacrificed between 1 and 28 days after transfer. Fig. 2Ashows that
after adoptive transfer, both CD4
and CD8
T cells persisted in
the spleen of CD3
/
recipient mice. Cell numbers increased
slowly up to day 3 and thereafter more rapidly. At day 28 post-
transfer, the CD3
/
spleen contained 15 10
6
CD4
and 5
10
6
CD8
T cells, respectively, or a 10- to 15-fold expansion of
FIGURE 1. IL-7 overexpression perturbs B cell lym-
phopoiesis. A, BM cells were three-color stained as de-
scribed in Materials and Methods. Bars show the aver-
age (n3) cell number (left ordinates) of the indicated
subpopulation. p, Ratio of each subpopulation in IL-
7Tg.CD3
/
vs CD3
/
mice (right ordinates). B,
Spleen cells were three-color stained with anti-CD19,
anti-B220, and anti-IgM or anti-CD19, anti-CD21, and
anti-CD23. Upper cytograms, B220 vs IgM; lower cy-
tograms, CD23 vs CD21 profiles of gated CD19
cells.
Right bar graph, Mean number and ratios of the indi-
cated subpopulations. Imm.B, Immature B cells; FB,
follicular B; MZB, marginal zone B. C, Thymus cells
were four-color stained with anti-CD25, anti-CD44, anti-
B220, and anti-IgM. Upper cytograms show CD25 vs
CD44 profiles of all live cells; lower cytograms show
the B220 vs IgM staining of cells gated for intermediate
CD44-expressing cells (round gate in upper cytograms).
Right bar graph shows mean number and ratios of the
indicated subpopulations. In each cytogram display,
numbers in quadrants represent the percentage of posi-
tive cells. Results are representative of at least five
experiments.
164 INCREASING IL-7 AVAILABILITY ALTERS T CELL HOMEOSTASIS
the transferred cells between days 1 and 28. Thus, numbers are
almost similar to those in the spleen of a normal B6 mouse.
Overexpression of IL-7 in CD3
/
mice had a significant effect
on the behavior of transferred T cells. Thus, we observed a greater
rate of expansion of both CD4
and CD8
T cells in IL-
7Tg.CD3
/
recipient mice and also an increase in long term cell
recovery. Therefore, from day 3 and up to day 28, IL-
7Tg.CD3
/
contained more CD4
and CD8
T cells than their
CD3
/
controls with lymphocyte numbers reaching a plateau at
about day 14. At day 28, the spleen of IL-7Tg.CD3
/
recipients
contained 52 10
6
CD4
and 17 10
6
CD8
T cells, repre-
senting a 3- to 4-fold higher expansion compared with control
CD3
/
recipients (Fig. 2A). A similar pattern of growth and
recovery was obtained in the LN (not shown). Thus, increased
availability of IL-7 in vivo increases the yield of T cells after
adoptive transfer.
Next, we investigated in more detail the in vivo impact of IL-7
overexpression on transferred T cells. To rule out a possible pref-
erential and exacerbated monoclonal expansion of T cells, we an-
alyzed their TCR
and TCR
repertoire by flow cytometry. The
repertoire of transferred T cells in both recipient mice was large,
polyclonal, and comparable with that of T cells from unmanipu-
lated B6 mice (not shown) with no obvious bias after expansion.
Then, we determined the phenotype of transferred T cells by
four-color flow cytometry. Analysis of T cells during early phases
of proliferation (days 3, 5, and 7 posttransfer) showed changes
similar to those in other published reports (3); namely, cells be-
came larger, as judged by forward light scatter, remained CD25
and CD69
and CD44
(not shown). One month after transfer,
CD4
and CD8
T cells had acquired a memory-like phenotype
(3, 39 42) (Fig. 2B, left and middle cytograms) with most CD4
T cells being CD44
high
CD62L
low
and many CD8
T cells being
CD44
high
CD122
high
. This phenotypic conversion that accompanies
expansion of polyclonal T cells in both recipient mice was only
slightly more pronounced in IL-7 Tg recipients. Additionally, we
observed that 10% of the injected CD4
T cells spontaneously
express CD25. Therefore, among CD4
cells, the proportion of
CD25
cells in both types of recipient mice remained the same
with 10% of CD4
T cells expressing CD25 at 28 days after
transfer (Fig. 2B, right cytograms).
Localization of transferred cells in vivo
It has been shown that transferred T cells migrate to the periarte-
riolar lymphocyte sheaths (PALS) of the spleen where they pre-
sumably receive division and survival signals (43). To exclude that
altered T cell recovery was due to a difference in spleen architec-
ture, a confocal immunohistology study was conducted. Despite
IL-7-dependent alteration of BM B cell development (Fig. 1B) and
the presence of immature B cells, no difference in B cell follicle
size or organization was observed in the spleen of IL-
7Tg.CD3
/
mice (Fig. 3). At day 1 posttransfer (Fig. 3, aand b),
CFSE
cells were found in PALS of both recipients, with slightly
more in the follicles of IL-7Tg.CD3
/
mice. At day 28 post-
transfer, T cells had lost CFSE labeling but were identified using
anti-CD90 (Thy-1) staining. Results indicated that transferred T
cells were localized to T cell areas of follicles but that restoration
of these areas in both recipients was only partial compared with B6
controls (Fig. 3, c– e). These findings suggest that T cells undergo
LIP within a dedicated area inside B cell follicles (43) that pre-
sumably provides specific factors, including IL-7, necessary for
maintaining T cell survival and proliferation.
IL-7 overexpression has differential proliferative effects on
CD8
and CD4
subpopulations
Using CFSE labeling, we compared the proliferation of T cells in
lymphopenic hosts differing in IL-7 availability. Three days after
transfer into CD3
/
recipients, few CD4
cells had divided (Fig.
4A, left panels) and the average number of divisions was below 1.
Over the same time period, CD8
T cells had proliferated more, with
an average of almost two divisions. In contrast, in IL-7Tg.CD3
/
recipients, a greater proportion of both CD4
and CD8
T cells had
reduced CFSE fluorescence and had undergone between one and four
rounds of division, respectively. That increasing IL-7 availability pro-
moted LIP of CD8
T cells was somewhat expected, but in contrast
to other published reports (6, 7), there was also a clear effect of IL-7
on CD4
T cells. At 5 days after transfer (Fig. 4A, right panels), T
cells had undergone more rounds of division. Again CD8
cells had
divided more than CD4
cells. This difference between CD8
and
CD4
cells was maintained regardless of differences in IL-7 avail-
ability. Both T cell subpopulations proliferated more in IL-
7Tg.CD3
/
vs CD3
/
littermates. Similar differences between
CD8
and CD4
cell division were also seen when the cells were
transferred to IL-7Tg.RAG-2
/
or RAG-2
/
littermates where the
difference in IL-7 transcript levels was 3- to 5-fold (not shown).
Again, because there was no up-regulation of CD69 or CD25 expres-
sion on either CD8
or CD4
T cells (not shown) this, together with
the relatively slow kinetics of proliferation, indicated that LIP rather
than activation-induced proliferation was being measured (39).
FIGURE 2. IL-7 enhances T cell recovery but not
memory phenotype conversion after adoptive transfer.
A, Upper graph, number (SD, n7) of recovered
CD4
cells (,f)(10
6
); lower graph, number of
CD8
cells (,Œ)(10
6
) recovered at various times
after transfer into CD3
/
(f,Œ) or IL-7Tg.CD3
/
(,) recipients. B, Fresh spleen cells from B6 mice (i)
or 28 days after transfer into CD3
/
(ii)orIL-
7Tg.CD3
/
(iii) mice were stained with either anti-
CD4, anti-CD25, anti-CD44, and anti-CD62L (left and
right) or anti-CD8, anti-CD44, and anti-CD122 (mid-
dle). Left cytograms, CD44 vs CD62L on live-gated
CD4
spleen cells; middle cytograms, CD44 vs CD122
on live-gated CD8
;right cytograms, CD4 vs CD25
distribution on live-gated spleen cells. The percentages
of cells within the indicated region are shown in each
dot plot (left and middle). In the right dot plots, indi-
cates the percent CD25
among total CD4
cells. Three
mice were individually examined in each group, and a
representative result is shown. FL, Fluorescence.
165The Journal of Immunology
After 14 days, the number of transferred T cells had reached a
plateau, and they became mostly CFSE negative. BrdU labeling
experiments showed that after 28 days, the turnover of transferred
T cells was similar in both recipients with 30 and 20% of CD4
and CD8
T cells, respectively, incorporating BrdU (Fig. 4B).
However, due to the overall increase in T cell recovery in IL-
7Tg.CD3
/
mice, (Fig. 2Aand data not shown), the total num-
ber of proliferating T cells was 4- to 5-fold higher in IL-7
transgenics.
In most experiments, we transferred unseparated populations of
T cells. However, to see whether naive T cells were equally af-
fected by differences in IL-7 availability, sorted naive
CD4
CD45RB
high
and CD8
CD44
low
cells were CFSE-labeled
and cotransferred into pairs of CD3
/
and IL-7Tg.CD3
/
mice. As shown in Fig. 5A, the CFSE profiles of transferred naive
CD4
and CD8
T cells showed that both responded to increased
IL-7 availability by an increase in the proportion of cells engaged
in LIP and the mean number of divisions (about one more division
for both CD4
and CD8
T cells in IL-7Tg recipients 4 days
posttransfer). Simultaneous phenotypic analysis showed that inde-
pendent of the host, there was an increase in CD44 expression by
transferred CD4
and CD8
T cells (Fig. 5B).
Previous experiments had suggested that during LIP, naive T
cells transiently acquired a memory-like phenotype (40). However,
subsequent experiments clearly demonstrated that this transient
phenotypic conversion was due to the de novo production of naive
T cells by the freshly recolonized thymus of irradiated recipient
mice (3, 41, 42). As shown in Fig. 6, the thymus of both CD3
/
and IL-7Tg.CD3
/
murine recipients contained neither
CD4
CD8
double-positive cells (Fig. 6, left panels) nor
CD44
CD25
(DN4) thymocytes (Fig. 6, right panels). This
shows the absence of thymus reconstitution by T cell progenitors
in unirradiated recipients and that the observed BrdU incorporation
into peripheral T cells (Fig. 4C) was not the result of de novo
FIGURE 3. Transferred CFSE
cells homed to
PALS following transfer. Frozen spleen sections were
stained as described in Materials and Methods. Recipi-
ent mice were killed 1 day (aand b)or4wk(cand d)
after transfer of CFSE-labeled pooled LN spleen
cells. Sections were stained for B cells (red) with PE-
conjugated anti-B220 mAb (a– e). CFSE
donor cells
(green) are situated almost exclusively in the periarte-
riolar lymphocyte sheath (PALS) (aand b). Four weeks
after transfer (cand d), transferred T cells had lost CFSE
staining, and T cells were stained with FITC-conjugated
anti-CD90.2 (Thy1.2) mAb. The T area is not com-
pletely restored (cand d) compared with normal B6
mice (e). Sections aand care from CD3
/
recipients,
and sections band dare from IL-7Tg.CD3
/
recipi-
ents. Data are representative of one of three mice ana-
lyzed individually. Scale bar, 80
m.
FIGURE 4. IL-7 promotes proliferation of poly-
clonal CD4
and CD8
cells in lymphopenic mice. A,
CFSE profiles of gated CD4
or CD8
spleen cells 3
days (left panels) or 5 days (right panels) after transfer
of 10
7
pooled labeled LN and spleen cells into CD3
/
(upper panels) or IL-7Tg.CD3
/
(bottom panels)re
-
cipients. Similar results were obtained with LN cells
(not shown). B, Cytoplasmic BrdU profiles of gated
CD4
(left histograms)orCD8
(right histograms)
cells 28 days after transfer and labeled for the last 5 days
with BrdU as outlined in Materials and Methods.
Within each panel, the percent of BrdU
cells is indi-
cated. Negative controls were 1% BrdU
. FL,
Fluorescence.
166 INCREASING IL-7 AVAILABILITY ALTERS T CELL HOMEOSTASIS
thymus T cell production. Surprisingly, more CD4
and CD8
single-positive (SP) cells were found in the thymus of IL-
7Tg.CD3
/
mice compared with CD3
/
recipients (Fig. 6,
left panels) and at early time points, these SP cells were CFSE
(not shown).
IL-7 decreases T cell apoptosis with a temporal effect on CD4
and CD8
T cells
Regulation of T cell survival is a critical feature for the mainte-
nance of peripheral T cell numbers. To determine whether increas-
ing IL-7 availability promoted cell survival during LIP, two pa-
rameters were measured, namely, annexin V staining and
cytoplasmic Bcl-2 expression. At 3 days after transfer, the propor-
tion of annexin V
CD4
T cells was significantly reduced in
spleens of IL-7Tg.CD3
/
mice, and CD4
T cell apoptosis was
reduced (20 40%) less for each round of division (Fig. 7A). At
this time, cytoplasmic Bcl-2 levels in CD4
T cells was already
elevated in transgenic recipients and this persisted until 28 days
posttransfer (Fig. 7C).
In contrast, the early antiapoptotic action of IL-7 on transferred
CD8
T cells was less pronounced (Fig. 7, Aand C). In IL-
7Tg.CD3
/
recipients, 3 days posttransfer, there was only a
5–10% reduction in apoptosis for each round of division (Fig. 7A)
and no increase in cytoplasmic Bcl-2 expression (Fig. 7C). How-
ever, by day 28, annexin V
cells had decreased (Fig. 7B), and
Bcl-2 levels had increased in CD8
T cells and were even higher
than in CD4
T cells (Fig. 7). Thus, the effects of increasing IL-7
availability have temporally independent effects on CD4
vs
CD8
T cells. All the changes described above in polyclonal pop-
ulations of CD8
lymphocytes were also observed with TCR-
transgenic cells. Thus, when CD8
T cells from P14.RAG-2
/
mice were transferred to IL-7Tg.CD3
/
or CD3
/
recipients,
proliferation was more rapid, apoptosis was decreased, expression
of cytoplasmic Bcl-2 was increased, and cell recovery at 28 days
increased 10-fold in IL-7Tg.CD3
/
compared with CD3
/
recipients (not shown).
The transferred CFSE-labeled cells contained B cells, but there
did not appear to be an effect of IL-7 overexpression on their
proliferation, survival, or Bcl-2 expression (Fig. 7Cand data not
shown) in either IL-7Tg.CD3
/
or CD3
/
recipients. In con-
trast, in completely lymphopenic RAG-2
/
or IL-7Tg.RAG-2
/
recipients, transferred B cells proliferated slowly but proliferation
was similar in IL-7 transgenic and nontransgenic recipients (N.
Bosco, unpublished observation).
Discussion
In this report, we have studied the behavior of polyclonal popula-
tions of lymphocytes transferred into novel T-lymphopenic IL-7-
transgenic recipient mice (Figs. 1 and 2). Results obtained indicate
that increasing IL-7 enhanced recovery of both CD4
and CD8
polyclonal T cells but had markedly different effects on prolifera-
tion (Figs. 4 and 5) and apoptosis (Fig. 7) of transferred cells
depending on whether cells were actively undergoing LIP or sur-
viving in a replenished peripheral compartment. The prediction
had been made that in lymphopenic recipients, increasing IL-7
availability in vivo should increase the rate of T cell proliferation
(27), but this hypothesis has not been directly addressed. Our in
vivo results clearly demonstrate that during LIP, increasing IL-7
availability increases the rate of division of both CD4
and CD8
T cells but not of B cells.
The rate of proliferation of CD8
T cells, as measured by CFSE
fluorescence, was enhanced by increasing IL-7 availability. This
was true for both polyclonal and P14 TCR-transgenic T cells. The
proliferation of CD4
cells, although intrinsically slower than that
FIGURE 6. Normal thymopoiesis is not restored after adoptive transfer
of polyclonal lymphocytes. One month after transfer, recipient thymuses
were recovered and stained with anti-CD4, anti-CD8, anti-CD25, and anti-
CD44. Shown are the CD4 vs CD8 cytograms on all (left) or CD25 vs
CD44 cytograms of gated CD4
/CD8
cells (right) from adult B6 (upper)
or adoptively transferred CD3
/
(middle) or IL-7Tg.CD3
/
(lower)
recipients. CD44
low
CD25
cells in recipient mice are B cells (see Fig. 1).
Left lower panels, Percentage of SP cells within the indicated regions.
Insets, Region used to define DN and SP cells.
FIGURE 5. Naive CD4
and CD8
T cells prolifer-
ate more rapidly in IL-7Tg.CD3
/
mice. A, CFSE
profiles of 4 10
6
sorted, pooled and labeled, naive
CD4
CD45RB
high
(left panels)orCD8
CD44
low
(right panels) T cells 4 days after transfer into CD3
/
(upper panels) or IL-7Tg.CD3
/
mice (lower panels).
Division numbers and percentages of nondivided cells
are indicated. B, CD44 expression level vs CFSE con-
tent of the indicated cells in the corresponding mice.
Data are representative of two independent experiments
with two mice analyzed individually. FL, Fluorescence.
167The Journal of Immunology
of CD8
cells, was also increased in IL-7Tg.CD3
/
recipients
(Figs. 4 and 5). Although initial results had indicated that prolif-
eration of CD4
SP neonatal thymocytes in vitro (44) could be
maintained in the presence of IL-7, subsequent reports had indi-
cated that IL-7 had no effect on CD4
T cell proliferation (6, 7),
but this notion has been recently revised (27, 45, 46).
In IL-7Tg.CD3
/
recipients, Bcl-2 was up-regulated in both
CD4
and CD8
T cells but with different kinetics (Fig. 7). Early
after transfer, there was a distinct effect of IL-7 availability on
Bcl-2 expression in CD4
cells whereas CD8
cells were not
affected. Later on, Bcl-2 expression and apoptosis resistance were
observed and were more pronounced in CD8
T cells. In contrast,
Bcl-2 levels in T cells transferred into CD3
/
recipients were
similar to those of unmanipulated B6 controls. It has been shown
that CD8
but not CD4
memory cells overexpress Bcl-2 protein
(47). Phenotypic analysis indicated that after LIP, in both trans-
genic and nontransgenic recipients, T cells were enriched in so-
called memory-like cells (3). Differences in the proportion of
memory-like cells within surviving cells were minor, yet there was
a clear increase in cytoplasmic Bcl-2 expression (Figs. 2 and 7).
This indicates that changes in Bcl-2 levels were IL-7 dependent
and not the result of an altered cellular composition of CD4
or
CD8
T cell subsets. Even so-called naive CD4
CD45RB
high
and
CD8
CD44
low
T cells respond to increased IL-7 availability (Fig.
5A) and undergo naive to memory-like phenotypic conversion af-
ter transfer into lymphopenic hosts (Fig. 5Band Refs. 3 and 48).
Although sharing certain properties with true memory cells, it is
generally agreed that the slower kinetics of induction of effector
functions by memory-like cells clearly distinguishes them from
true memory cells (48 –50). It could be either that IL-7 sustains
naive T cell differentiation into memory cells or that IL-7 promotes
selection of memory CD8
subset among the transferred cells that
then outcompete naive T cells in LIP. The latter hypothesis would
be in agreement with our previous report describing an increase of
naturally arising CD8
memory cells in nonimmunized IL-7Tg.B6
mice (26). Importantly, the combined effects of decreasing annexin
V staining and increasing cytoplasmic Bcl-2 expression could ac-
count for the increased T cell yield in IL-7-transgenic recipients.
This reproducible difference in the degree of apoptosis is important
if we consider the exponential growth of T cells during LIP. De-
creasing cell loss by 10 or 15% at each division can lead to a 50%
increase in cell recovery after eight or five divisions, respectively.
Indeed, cell recovery is a balance between the rate of cell prolif-
eration and cell loss. The inoculum used in most of our experi-
ments contained twice as many CD4 as CD8 T cells, and this
difference may partially explain the increased recovery of CD4
vs CD8
T cells 1 month after transfer. In addition, as shown in
Fig. 7, there is a major effect of IL-7 overexpression on Bcl-2
expression by CD4
but not CD8
T cells early after transfer.
This is likely to have major consequences on overall cell recovery.
This could explain why CD4
T cell recovery is higher than CD8
cells despite their slower proliferation rate.
One month after transfer, when the T cell compartment had
reached equilibrium, there was no difference in overall turnover of
CD4
or CD8
T cells, whereas the total number of T cells was
4 times higher in IL-7Tg.CD3
/
recipients (Figs. 2 and 4). That
increasing IL-7 availability per se does not increase turnover of the
established T cell pool is consistent with studies in IL-7Tg.B6
mice (26) (N. Bosco, unpublished observation) in which the pro-
portion of dividing CD4
or CD8
T cells is similar to that of
FIGURE 7. Kinetics of Bcl-2 up-regulation and survival by IL-7 differs between CD4
and CD8
cells. A, Bars show the mean percent (SD, n
3) of annexin V
CD4
(upper panels)orCD8
(lower panels) cells that had divided the indicated number of times 3 days posttransfer into CD3
/
(f)
or IL-7Tg.CD3
/
() recipients. Insets, CFSE vs annexin V cytograms of the corresponding cells in CD3
/
recipients. B, Bar graphs show the mean
(SD, n3) of apoptotic (annexin V
) gated CD4
(left)orCD8
(right) cells 28 days posttransfer into CD3
/
(f) or IL-7Tg.CD3
/
()
recipients. Cells were stained with anti-CD4, anti-CD8, anti-annexin V, and 7-aminoactinomycin D as described in Materials and Methods.C, Bar graphs
show the mean fluorescence intensity (MFI) of intracellular Bcl-2 expression in B220
, CD4
,orCD8
lymphocytes from WT (f) or in cells 3 days (upper
panels) or 28 days (lower panels) after transfer into CD3
/
(u) or IL-7Tg.CD3
/
() recipients. Four mice were individually examined in each group,
and the mean fluorescence intensity SD is represented. WT, Wild type.
168 INCREASING IL-7 AVAILABILITY ALTERS T CELL HOMEOSTASIS
nontransgenic controls. However, given the increased pool size of
T cells, the number of dividing cells is proportionately higher, and
equilibrium at this elevated cell number is maintained by balanced
proliferation and simultaneous cell loss.
The IL-7R signal transduction cascade has been shown to acti-
vate expression of Bcl-2 family genes (51). We showed a temporal
difference in Bcl-2 expression between CD4
and CD8
T cells in
response to increased availability of IL-7 (Fig. 7). As recently
reported (52), the signaling cascade downstream of the IL-7R may
be differentially regulated between CD4
and CD8
cells. IL-7R
gene transcription is suppressed in response to IL-7 signaling, a
process that involves different molecular mechanisms in CD4
vs
CD8
T cells. CD8
T cells use the transcriptional repressor
GFI1; the transcriptional repressor used by CD4
T cells remains
to be identified. If major differences exist in the repertoire of tran-
scription activators and repressors used by different T cell subsets
after IL-7R engagement, these factors could constitute important
targets to control selectively T cell responses to various cytokines
and therefore T cell homeostasis.
T cells injected into lymphopenic animals migrated into lym-
phoid follicles, and reconstituted their T cell areas. Restricted
homing of transferred T cells into lymphoid organs suggests that
lymphocyte migratory properties are crucial for the initiation of
LIP. Pertussis toxin treatment of donor cells abrogates G protein-
dependent migration of T cells and reduces LIP in recipient mice
(43). IL-7 and additional resources, including chemokines, are
present inside lymphoid follicles, and the concentration of these
factors is presumably increased in lymphopenic conditions (23).
This may be how T cells sense lymphopenia and how LIP is trig-
gered. Experiments in vitro have shown that IL-7 decreases the
activation threshold of T cells, thereby serving as a “cofactor” for
activation (45). Our results confirm that a similar mechanism
might be operational in vivo, thereby promoting LIP. The cells that
deliver the signals promoting LIP remain to be defined. However,
dendritic cells are good candidates because some of them localize
inside lymphoid follicles, express MHC molecules, secrete cyto-
kines, and sustain LIP in vitro (53).
Where does T cell proliferation during LIP occur? Most trans-
ferred cells localize to the PALS of lymphoid follicles (Fig. 3), the
anatomical site where most proliferation occurs (43). However, it
is unclear whether cells remain fixed in the spleen during LIP.
Early after cell transfer, CFSE
CD4
and CD8
SP cells were
found in the thymus of recipients, and their CFSE profiles mirrored
those of peripheral T cells. Quantitative RT-PCR and analysis of
thymic B cell phenotype together indicated that the thymus was
enriched in IL-7 (Fig. 1Cand N. Bosco, unpublished observation).
That CFSE
T cells were present in the thymus indicated that their
survival could be maintained in anatomical sites rich in IL-7. Their
presence in the thymus could also indicate that cells circulate dur-
ing LIP. A homeostatic niche supporting T cell LIP could be sim-
ply defined as a location where the trophic factors are available.
These locations for T cells are somehow flexible, a situation anal-
ogous to that described for B cells by Agene`s and Freitas (54).
Transfer of B lymphocytes into IL-Tg.CD3
/
or IL-7Tg.Rag-
2
/
recipients indicated that IL-7 was not involved in B cell LIP
(not shown). This is consistent with our previous data using IL-
7Tg.B6 mice, where the increase in mature B cell compartments is
simply a consequence of increasing BM B lymphopoiesis and not
due to a role for IL-7 in mature B cell survival (32). Thus, as
previously reported, T and B cell homeostasis are independent of
one another and depend on different resources (55).
In conclusion, we describe for the first time the pleiotropic ef-
fects of IL-7 during and after lymphocyte polyclonal LIP. We
show that increasing IL-7 availability has effects on both CD4
and CD8
T cells but not on B cells. These results are relevant to
the clinical settings in which IL-7 is being used as an adjunct for
hemopoietic, in particular lymphocyte reconstitution, after BM
transfer (10). Much less information is available on the role of IL-7
in human T cell LIP, but recent advances, including the generation
of humanized RAG
/
.
c
/
mice reconstituted with human cord
blood cells (56) could provide opportunities to explore the effect of
human IL-7 on adoptively transferred human lymphocytes.
Acknowledgments
We thank Patrice Marche and Institut National de la Sante´etdelaRe-
cherche Me´dicale for their support; Dr. Jo¨ rg Kirberg, Max Planck Institute
for Immunobiology, Freiburg for P14.RAG-2
/
mice; Dr. Ton Rolink for
invaluable help; and Drs. Simon Fillatreau and Didier Grunwald for con-
focal microscopy. We thank Eve Borel for mouse maintenance and
Ve´ronique Collin for cell sorting. We thank Drs. Serge Cande´ias and Chris-
tophe Viret for their comments and constructive criticisms of the
manuscript.
Disclosures
The authors have no financial conflict of interest.
References
1. Marrack, P., and J. Kappler. 2004. Control of T cell viability. Annu. Rev. Immu-
nol. 22: 765–787.
2. Jameson, S. C. 2002. Maintaining the norm: T-cell homeostasis. Nat. Rev. Im-
munol. 2: 547–556.
3. Ge, Q., H. Hu, H. N. Eisen, and J. Chen. 2002. Naive to memory T-cell differ-
entiation during homeostasis-driven proliferation. Microbes Infect. 4: 555–558.
4. Sprent, J., and C. D. Surh. 2003. Cytokines and T cell homeostasis. Immunol.
Lett. 85: 145–149.
5. Ceredig, R. 2000. Interleukin-7, a non-redundant potent cytokine whose over-
expression massively perturbs B-lymphopoiesis. In Cytokines and Cytokine Re-
ceptors. C. A. Bona and J.-P. Revillard, eds. Harwood Academic Publishers, pp.
167–182.
6. Vella, A. T., S. Dow, T. A. Potter, J. Kappler, and P. Marrack. 1998. Cytokine-
induced survival of activated T cells in vitro and in vivo. Proc. Natl. Acad. Sci.
USA 95: 3810–3815.
7. Tan, J. T., E. Dudl, E. LeRoy, R. Murray, J. Sprent, K. I. Weinberg, and
C. D. Surh. 2001. IL-7 is critical for homeostatic proliferation and survival of
naive T cells. Proc. Natl. Acad. Sci. USA 98: 8732–8737.
8. Morrissey, P. J., P. Conlon, K. Charrier, S. Braddy, A. Alpert, D. Williams,
A. E. Namen, and D. Mochizuki. 1991. Administration of IL-7 to normal mice
stimulates B-lymphopoiesis and peripheral lymphadenopathy. J. Immunol. 147:
561–568.
9. Alpdogan, O., S. J. Muriglan, J. M. Eng, L. M. Willis, A. S. Greenberg,
B. J. Kappel, and M. R. Van Den Brink. 2003. IL-7 enhances peripheral T cell
reconstitution after allogeneic hematopoietic stem cell transplantation. J. Clin.
Invest. 112: 1095–1107.
10. Fry, T. J., and C. L. Mackall. 2002. Interleukin-7: from bench to clinic. Blood 99:
3892–3904.
11. Ferrara, J. L. 1993. Cytokine dysregulation as a mechanism of graft versus host
disease. Curr. Opin. Immunol. 5: 794–799.
12. Moses, C. T., K. M. Thorstenson, S. C. Jameson, and A. Khoruts. 2003. Com-
petition for self ligands restrains homeostatic proliferation of naive CD4 T cells.
Proc. Natl. Acad. Sci. USA 100: 1185–1190.
13. Chung, B., L. Barbara-Burnham, L. Barsky, and K. Weinberg. 2001. Radiosen-
sitivity of thymic interleukin-7 production and thymopoiesis after bone marrow
transplantation. Blood 98: 1601–1606.
14. Kieper, W. C., J. T. Burghardt, and C. D. Surh. 2004. A role for TCR affinity in
regulating naive T cell homeostasis. J. Immunol. 172: 40– 44.
15. Kassiotis, G., R. Zamoyska, and B. Stockinger. 2003. Involvement of avidity for
major histocompatibility complex in homeostasis of naive and memory T cells.
J. Exp. Med. 197: 1007–1016.
16. Ge, Q., A. Bai, B. Jones, H. N. Eisen, and J. Chen. 2004. Competition for self-
peptide-MHC complexes and cytokines between naive and memory CD8
T
cells expressing the same or different T cell receptors. Proc. Natl. Acad. Sci. USA
101: 3041–3046.
17. Hori, S., M. Haury, A. Coutinho, and J. Demengeot. 2002. Specificity require-
ments for selection and effector functions of CD25
4
regulatory T cells in
anti-myelin basic protein T cell receptor transgenic mice. Proc. Natl. Acad. Sci.
USA 99: 8213–8218.
18. Apostolou, I., A. Sarukhan, L. Klein, and H. von Boehmer. 2002. Origin of
regulatory T cells with known specificity for antigen. Nat. Immunol. 3: 756 –763.
19. Almeida, A. R., N. Legrand, M. Papiernik, and A. A. Freitas. 2002. Homeostasis
of peripheral CD4
T cells: IL-2R
and IL-2 shape a population of regulatory
cells that controls CD4
T cell numbers. J. Immunol. 169: 4850– 4860.
20. Stockinger, B., G. Kassiotis, and C. Bourgeois. 2004. Homeostasis and T cell
regulation. Curr. Opin. Immunol. 16: 775–779.
169The Journal of Immunology
21. Annacker, O., O. Burlen-Defranoux, R. Pimenta-Araujo, A. Cumano, and
A. Bandeira. 2000. Regulatory CD4 T cells control the size of the peripheral
activated/memory CD4 T cell compartment. J. Immunol. 164: 3573–3580.
22. Banz, A., A. Peixoto, C. Pontoux, C. Cordier, B. Rocha, and M. Papiernik. 2003.
A unique subpopulation of CD4
regulatory T cells controls wasting disease,
IL-10 secretion and T cell homeostasis. Eur. J. Immunol. 33: 2419–2428.
23. Fry, T. J., and C. L. Mackall. 2001. Interleukin-7: master regulator of peripheral
T-cell homeostasis? Trends Immunol. 22: 564–571.
24. Fisher, A. G., C. Burdet, M. LeMeur, D. Haasner, P. Gerber, and R. Ceredig.
1993. Lymphoproliferative disorders in an IL-7 transgenic mouse line. Leukemia
7 (Suppl. 2): S66–S68.
25. Mertsching, E., C. Burdet, and R. Ceredig. 1995. IL-7 transgenic mice: analysis
of the role of IL-7 in the differentiation of thymocytes in vivo and in vitro. Int.
Immunol. 7: 401–414.
26. Kieper, W. C., J. T. Tan, B. Bondi-Boyd, L. Gapin, J. Sprent, R. Ceredig, and
C. D. Surh. 2002. Overexpression of interleukin (IL)-7 leads to IL-15-indepen-
dent generation of memory phenotype CD8
T cells. J. Exp. Med. 195:
1533–1539.
27. Li, J., G. Huston, and S. L. Swain. 2003. IL-7 promotes the transition of CD4
effectors to persistent memory cells. J. Exp. Med. 198: 1807–1815.
28. Malissen, M., A. Gillet, L. Ardouin, G. Bouvier, J. Trucy, P. Ferrier, E. Vivier,
and B. Malissen. 1995. Altered T cell development in mice with a targeted mu-
tation of the CD3-
gene. EMBO J. 14: 4641–4653.
29. Shinkai, Y., G. Rathbun, K. P. Lam, E. M. Oltz, V. Stewart, M. Mendelsohn,
J. Charron, M. Datta, F. Young, A. M. Stall, et al. 1992. RAG-2-deficient mice
lack mature lymphocytes owing to inability to initiate V(D)J rearrangement. Cell
68: 855–867.
30. Pircher, H., K. Burki, R. Lang, H. Hengartner, and R. M. Zinkernagel. 1989.
Tolerance induction in double specific T-cell receptor transgenic mice varies with
antigen. Nature 342: 559–561.
31. Agenes, F. 2003. B lymphocyte life span, rate of division and differentiation are
regulated by total cell number. Eur. J. Immunol. 33: 1063–1069.
32. Ceredig, R., N. Bosco, P. N. Maye, J. Andersson, and A. Rolink. 2003. In in-
terleukin-7-transgenic mice, increasing B lymphopoiesis increases follicular but
not marginal zone B cell numbers. Eur. J. Immunol. 33: 2567–2576.
33. Laurent, J., N. Bosco, P. N. Marche, and R. Ceredig. 2004. New insights into the
proliferation and differentiation of early mouse thymocytes. Int. Immunol. 16:
1069 –1080.
34. Hayakawa, K., D. Tarlinton, and R. R. Hardy. 1994. Absence of MHC class II
expression distinguishes fetal from adult B lymphopoiesis in mice. J. Immunol.
152: 4801–4807.
35. Mertsching, E., U. Grawunder, V. Meyer, T. Rolink, and R. Ceredig. 1996. Phe-
notypic and functional analysis of B lymphopoiesis in interleukin-7- transgenic
mice: expansion of pro/pre-B cell number and persistence of B lymphocyte de-
velopment in lymph nodes and spleen. Eur. J. Immunol. 26: 28–33.
36. Ceredig, R., J. Andersson, F. Melchers, and A. Rolink. 1999. Effect of deregu-
lated IL-7 transgene expression on B lymphocyte development in mice express-
ing mutated pre-B cell receptors. Eur. J. Immunol. 29: 2797–2780.
37. Ceredig, R. 2002. The ontogeny of B cells in the thymus of normal, CD3e knock-
out (KO), RAG-2 KO and IL-7 transgenic mice. Int. Immunol. 14: 87–99.
38. Ceredig, R., and T. Rolink. 2002. Opinion: a positive look at double-negative
thymocytes. Nat. Rev. Immunol. 2: 888– 897.
39. Surh, C. D., and J. Sprent. 2000. Homeostatic T cell proliferation: how far can T
cells be activated to self-ligands? J. Exp. Med. 192: F9–F14.
40. Goldrath, A. W., L. Y. Bogatzki, and M. J. Bevan. 2000. Naive T cells transiently
acquire a memory-like phenotype during homeostasis-driven proliferation.
J. Exp. Med. 192: 557–564.
41. Tanchot, C., A. Le Campion, B. Martin, S. Leaument, N. Dautigny, and B. Lucas.
2002. Conversion of naive T cells to a memory-like phenotype in lymphopenic
hosts is not related to a homeostatic mechanism that fills the peripheral naive T
cell pool. J. Immunol. 168: 5042–5046.
42. Ge, Q., H. Hu, H. N. Eisen, and J. Chen. 2002. Different contributions of thy-
mopoiesis and homeostasis-driven proliferation to the reconstitution of naive and
memory T cell compartments. Proc. Natl. Acad. Sci. USA 99: 2989–2994.
43. Dummer, W., B. Ernst, E. LeRoy, D. Lee, and C. Surh. 2001. Autologous reg-
ulation of naive T cell homeostasis within the T cell compartment. J. Immunol.
166: 2460–2468.
44. Ceredig, R., and C. Waltzinger. 1990. Neonatal mouse CD4
mature thymocytes
show responsiveness to interleukin 2 and interleukin 7: growth in vitro of neg-
atively selected V
6- and V
11-expressing CD4
cells from (C57BL/6 DBA/
2)F
1
mice. Int. Immunol. 2: 869– 877.
45. Seddon, B., and R. Zamoyska. 2003. Regulation of peripheral T-cell homeostasis
by receptor signalling. Curr. Opin. Immunol. 15: 321–324.
46. Kondrack, R. M., J. Harbertson, J. T. Tan, M. E. McBreen, C. D. Surh, and
L. M. Bradley. 2003. Interleukin 7 regulates the survival and generation of mem-
ory CD4 cells. J. Exp. Med. 198: 1797–1806.
47. Grayson, J. M., A. J. Zajac, J. D. Altman, and R. Ahmed. 2000. Cutting edge:
increased expression of Bcl-2 in antigen-specific memory CD8
T cells. J. Im-
munol. 164: 3950–3954.
48. Goldrath, A. W., C. J. Luckey, R. Park, C. Benoist, and D. Mathis. 2004. The
molecular program induced in T cells undergoing homeostatic proliferation.
Proc. Natl. Acad. Sci. USA 101: 16885–16890.
49. Kaech, S. M., J. T. Tan, E. J. Wherry, B. T. Konieczny, C. D. Surh, and
R. Ahmed. 2003. Selective expression of the interleukin 7 receptor identifies
effector CD8 T cells that give rise to long-lived memory cells. Nat. Immunol. 4:
1191–1198.
50. Seder, R. A., and R. Ahmed. 2003. Similarities and differences in CD4
and
CD8
effector and memory T cell generation. Nat. Immunol. 4: 835– 842.
51. Kovanen, P. E., and W. J. Leonard. 2004. Cytokines and immunodeficiency dis-
eases: critical roles of the
-dependent cytokines interleukins 2, 4, 7, 9, 15, and
21, and their signaling pathways. Immunol. Rev. 202: 67–83.
52. Park, J. H., Q. Yu, B. Erman, J. S. Appelbaum, D. Montoya-Durango,
H. L. Grimes, and A. Singer. 2004. Suppression of IL7R
transcription by IL-7
and other prosurvival cytokines; a novel mechanism for maximizing IL-7-depen-
dent T cell survival. Immunity 21: 289–302.
53. Ge, Q., D. Palliser, H. N. Eisen, and J. Chen. 2002. Homeostatic T cell prolif-
eration in a T cell-dendritic cell coculture system. Proc. Natl. Acad. Sci. USA 99:
2983–2988.
54. Agenes, F., and A. A. Freitas. 1999. Transfer of small resting B cells into im-
munodeficient hosts results in the selection of a self-renewing activated B cell
population. J. Exp. Med. 189: 319–330.
55. Freitas, A. A., and B. Rocha. 2000. Population biology of lymphocytes: the flight
for survival. Annu. Rev. Immunol. 18: 83–111.
56. Traggiai, E., L. Chicha, L. Mazzucchelli, L. Bronz, J. C. Piffaretti,
A. Lanzavecchia, and M. G. Manz. 2004. Development of a human adaptive
immune system in cord blood cell-transplanted mice. Science 304: 104–107.
170 INCREASING IL-7 AVAILABILITY ALTERS T CELL HOMEOSTASIS
... During thymocyte development, two signaling pathways, peptide presentation by major histocompatibility complex (MHC) molecules to T cell receptors (TCRs) and interleukin-7 (IL-7) signaling, are known to be critical for producing T cells in the thymus (2,3), but whether these pathways are involved in T cell survival in the periphery is debated. For example, although IL-7 is essential for thymic selection (2), its role in maintaining peripheral T cells is unclear, with some reports describing an essential role for IL-7 in T cell maintenance (4) and others showing that IL-7 is, by itself, unable to support peripheral T cell survival (5). Furthermore, whereas some studies have suggested an important role for peptideloaded MHC (pMHC)-mediated TCR activation for peripheral T cell expansion and survival (6), different studies, in part using the same transgenic mouse models, have reported that pMHC triggering of TCRs is dispensable for peripheral T cell survival (7,8). ...
... The other pathway besides pMHC:TCR signaling that has been implicated in peripheral naïve T cell survival is IL-7 receptor activation (4,30), although in a number of studies, IL-7 has also been found to be dispensable for T cell proliferation and survival (5,31). Activation of the IL-7 receptor results in the activation of Janus kinases (JAKs) and the signal transducer and activator of transcription 5 (Stat5) to induce the transcription of prosurvival genes, most notably the B cell lymphoma 2 (bcl2) gene (32). ...
Article
The control of T cell survival is crucial for defense against infectious pathogens or emerging cancers. Although the survival of peripheral naïve T cells has been proposed to be controlled by interleukin-7 (IL-7) signaling and T cell receptor (TCR) activation by peptide-loaded major histocompatibility complexes (pMHC), the essential roles for these pathways in thymic output and T cell proliferation have complicated the analysis of their contributions to T cell survival. Here, we showed that the WD repeat–containing protein coronin 1, which is dispensable for thymic selection and output, promoted naïve T cell survival in the periphery in a manner that was independent of TCR and IL-7 signaling. Coronin 1 was required for the maintenance of the basal activity of phosphoinositide 3-kinase δ (PI3Kδ), thereby suppressing caspase 8–mediated apoptosis. These results therefore reveal a coronin 1–dependent PI3Kδ pathway that is independent of pMHC:TCR and IL-7 signaling and essential for peripheral T cell survival.
... It is understood that homeostatic cytokines such as IL-7 and IL-15 play critical roles in the process of HP (3,5). Because T cells only consume and do not produce IL-7 or IL-15 (6,7), the availability of homeostatic cytokines is primarily controlled by the amount of cytokines produced by non-T cells and by the rate in which T cells consume these cytokines (8,9). Consequently, an increase in T cell numbers would result in decreased availability of homeostatic cytokines because the consumption of homeostatic cytokines would be increased. ...
... Consequently, an increase in T cell numbers would result in decreased availability of homeostatic cytokines because the consumption of homeostatic cytokines would be increased. However, a decrease in T cell numbers would conversely increase the in vivo availability of IL-7 and IL-15, resulting in the expansion of the peripheral T cell pool (6,8,10,11). In extreme cases, in which T cells are abolished, such as in chronic lymphopenic Rag-deficient mice or upon acute loss of T cells by sublethal irradiation, the amount of homeostatic cytokines is elevated to a level that they can initiate and drive the proliferation of adoptively transferred T cells (1,3,4). ...
Article
Lymphopenia-induced homeostatic proliferation (LIP) is a critical mechanism for restoring T cell immunity upon lymphodepleting insults or infections. LIP is primarily driven by homeostatic cytokines, such as IL-7 and IL-15, but not all T cells respond with the same efficiency to homeostatic proliferative cues. Although CD8 T cells vigorously proliferate under lymphopenic conditions, naive CD4 T cells are substantially impaired in their response to homeostatic cytokines, and they fail to fully expand. In this study, we show that the availability of IL-2Rβ (CD122), which is a receptor subunit shared by IL-2 and IL-15, affects both the cytokine responsiveness and the LIP of naive CD4 T cells in the mouse. The enumeration of surface IL-2Rβ molecules on murine naive CD4 and naive CD8 T cells revealed a 5-fold difference in IL-2Rβ abundance. Notably, it was the limited availability of IL-2Rβ that impaired CD4 T cell responsiveness to IL-15 and suppressed their LIP. As such, forced IL-2Rβ expression on CD4 T cells by transgenesis bestowed IL-15 responsiveness onto naive CD4 T cells, which thus acquired the ability to undergo robust LIP. Collectively, these results identify IL-2Rβ availability as a new regulatory mechanism to control cytokine responsiveness and the homeostatic proliferation of murine CD4 T cells.
... For example, the presence of CD4 T cells within the bone marrow or hematopoietic stem cell preparations correlates with better rate of thymopoiesis in bone marrow transplant recipients (14,23). In addition, animal studies showed that T cells can traffic from the periphery into the thymus and influence positive and negative thymocyte selection (24)(25)(26)(27)(28)(29). However, the mechanisms and consequences of such recirculation are still poorly understood and have not been examined under lymphopenic conditions. ...
... These findings in mATG-treated heart allograft recipients are consistent with the previous report by Sener et al that na€ ıve and memory CD4 T cell reconstitution is delayed in thymectomized nontransplanted mice injected with mATG or anti-lymphocyte serum (50). Mature CD4 and CD8 T cells can reenter the thymus and may be involved in negative selection by regulating the expression of tissue-specific antigens in the thymus and direct killing of antigen-specific T cells (24,25,(27)(28)(29)51). On the other hand, recirculating T cells have been implicated in thymocyte-positive selection (24). ...
Article
Full-text available
Lymphoablation is routinely used in transplantation, and its success is defined by the balance of pathogenic versus protective T cells within reconstituted repertoire. While homeostatic proliferation and thymopoiesis may both cause T cell recovery during lymphopenia, the relative contributions of these mechanisms remain unclear. The goal of this study was to investigate the role of the thymus during T cell reconstitution in adult allograft recipients subjected to lymphoablative induction therapy. Compared to euthymic mice, thymectomized heart allograft recipients demonstrated severely impaired CD4 and CD8 T cell recovery and prolonged heart allograft survival after lymphoablation with murine anti-thymocyte globulin (mATG). The injection with agonistic anti-CD40 mAb or thymus transplantation only partially restored T cell reconstitution in mATG treated thymectomized mice. Following mATG depletion, residual CD4 T cells migrated into the thymus following lymphoablation and enhanced thymopoiesis. Conversely, depletion of CD4 T cells prior to lymphoablation inhibited thymopoiesis at the stage of CD4(-) CD8(-) CD44(hi) CD25(+) immature thymocytes. This is the first demonstration that the thymus and peripheral CD4 T cells cooperate to ensure optimal T cell reconstitution following lymphoablation. Targeting thymopoiesis through manipulating functions of depletion-resistant helper T cells may thus improve therapeutic benefits and minimize risks of lymphoablation in clinical settings. This article is protected by copyright. All rights reserved.
... Nonetheless, the increased protein abundance of IL-7 can substantially amplify the prosurvival and proliferative effects of IL-7 as demonstrated in several in vivo studies. As such, the overexpression of IL-7 reportedly caused lymphoproliferative disorders with B cell hyperplasia and ectopic B cell development in mice, but could also induce chronic colitis depending on the site of transgenic IL-7 overexpression (31)(32)(33). In this regard, a recent study circumvented these issues by generating a tamoxifen-inducible IL-7 transgene where it was found that the increased IL-7 expression led to the dramatic expansion of memory phenotype CD8 T cells that was concomitant to a decrease in Foxp3 + Treg cells (34). ...
Article
Full-text available
The cytokine IL-7 plays critical and nonredundant roles in T cell immunity so that the abundance and availability of IL-7 act as key regulatory mechanisms in T cell immunity. Importantly, IL-7 is not produced by T cells themselves but primarily by non-lymphoid lineage stromal cells and epithelial cells that are limited in their numbers. Thus, T cells depend on cell extrinsic IL-7, and the amount of in vivo IL-7 is considered a major factor in maximizing and maintaining the number of T cells in peripheral tissues. Moreover, IL-7 provides metabolic cues and promotes the survival of both naïve and memory T cells. Thus, IL-7 is also essential for the functional fitness of T cells. In this regard, there has been an extensive effort trying to increase the protein abundance of IL-7 in vivo, with the aim to augment T cell immunity and harness T cell functions in anti-tumor responses. Such approaches started under experimental animal models, but they recently culminated into clinical studies, with striking effects in re-establishing T cell immunity in immunocompromised patients, as well as boosting anti-tumor effects. Depending on the design, glycosylation, and the structure of recombinantly engineered IL-7 proteins and their mimetics, recombinant IL-7 molecules have shown dramatic differences in their stability, efficacy, cellular effects, and overall immune functions. The current review is aimed to summarize the past and present efforts in the field that led to clinical trials, and to highlight the therapeutical significance of IL-7 biology as a master regulator of T cell immunity.
... The findings of our study may imply opportunities for the development of novel strategies in patient management and follow-up. In particular, novel therapies such as IL-7 immunotherapy have been previously explored in animal models to enhance lymphocyte recovery after treatment-induced lymphopenia [29,30]. Another potential strategy includes the reinfusion of autologous lymphocytes combined with GM-CSF to aid lymphocyte recovery after chemotherapy [31]. ...
Article
Full-text available
Hodgkin’s lymphoma carries an excellent prognosis with modern chemotherapy, but a significant proportion of patients remain refractory to or relapse after first-line treatment. Immunological changes post-treatment, such as chemotherapy-induced neutropenia (CIN) or lymphopenia, have shown prognostic significance in multiple tumor types. Our study aims to investigate the prognostic value of immunologic changes in Hodgkin’s lymphoma by examining the post-treatment lymphocyte count (pALC), neutrophil count (pANC) and the neutrophil-lymphocyte ratio (pNLR). Patients treated for classical Hodgkin’s lymphoma at the National Cancer Centre Singapore using ABVD-based regimens were retrospectively analyzed. An optimal cut-off value for high pANC, low pALC and high pNLR in predicting progression-free survival was determined by receiver operating curve analysis. Survival analysis was performed using the Kaplan–Meier method and multivariable Cox proportional models. Overall OS and PFS were excellent, with a 5-year OS of 99.2% and a 5-year PFS of 88.2%. Poorer PFS was associated with high pANC (HR 2.99, p = 0.0392), low pALC (HR 3.95, p = 0.0038) and high pNLR (p = 0.0078). In conclusion, high pANC, low pALC and high pNLR confer a poorer prognosis for Hodgkin’s lymphoma. Future studies should evaluate the potential of improving treatment outcomes by the adjustment of chemotherapy dose intensity based on post-treatment blood counts.
... Interleukin 7 (IL-7) is a haematopoietic growth factor protein that is secreted by stromal cells in the thymus and bone marrow [1][2][3]. IL-7 is one of the essential cytokines for the homoeostatic maintenance and development of B and T lymphocytes [4,5]. Its signalling cascade starts with IL-7 binding to the IL-7 receptor, which is composed of common gamma chain receptor (γc) and IL-7 receptor alpha (IL-7Rα) [6,7]. ...
Article
Full-text available
Interleukin 7 (IL-7) is an essential cytokine that acts as a potent growth factor of T-cells and supports the growth of B-cell precursors. IL-7 binds to a heterodimeric receptor consisting of an IL-7 receptor (alpha IL-7Rα) and the common gamma chain receptor (γc) which is shared with IL-2, IL-4, IL-9, IL-15 and IL21. The discovery of small-molecule agonists of cytokines would be of great pharmaceutical interest with the increasing scientific rationale. In this study, a series of molecular modelling methods, including field-based pharmacophore virtual screening, protein-protein docking and molecular dynamics simulations, led to the identification of two compounds (i.e. 1 and 2) of different classes that exhibit enhanced agonistic effects by activating the IL-7 signalling cascade. One of these compounds was selected as a hit and represents the first small-molecule agonist of IL-7Rα with single-digit micromolar activity. Moreover, the prediction model of the active compound to the IL-7Rα/γc interaction complex provides insight into the binding of a small-molecule agonist to its receptor.
... However, in Jak3 -/hosts, while WT 2C donor cells showed strong proliferative responses, Cd122 -/-2C donor cells failed to proliferate ( Figure 2B, top right two, and Supplementary Figure 2). When this adoptive transfer experiment was repeated in the same but irradiated (700 rad) hosts, both WT and Cd122 -/-2C donors showed similar degrees of slow LIP in control Jak3 +/hosts ( Figure 2B, bottom left two), which is known to be IL-7 (and self)dependent (26)(27)(28)(29). However, in irradiated Jak3 -/hosts, WT 2C donor cells showed markedly faster and greater proliferative responses than Cd122 -/-2C donor cells showing only slow rate of LIP ( Figure 2B, bottom right two, and Supplementary Figure 2), suggesting a role of CD122-dependent cytokines other than IL-7. ...
Article
Full-text available
The antigen-independent, strong proliferative responses of naive CD8⁺ T cells have been well demonstrated in a particular strain of mice lacking IL-2 receptors. This type of proliferation is mainly driven by common gamma-chain (γc) cytokines, such as IL-2, IL-7, and IL-15, present at abnormally high levels in these mice. Similarly, in the present study, we showed that mice lacking Janus kinase 3 (Jak3), a tyrosine kinase crucial for γc cytokine signaling, could induce strong proliferation of adoptively transferred naive CD8⁺ T cells. This proliferation was also independent of antigenic stimulation, but heavily dependent on IL-2, as evidenced by the failure of proliferation of adoptively transferred IL-2 receptor alpha- and beta-chain-deficient naive CD8⁺ T cells. Consistent with this, Jak3 –/– mice showed elevated serum levels of IL-2 compared to wild-type mice, and interestingly, IL-2 production was due to high levels of accumulation of activated CD4⁺ T cells in Jak3 –/– mice along with defective CD4⁺ T regulatory cells. Collectively, these findings reveal previously unidentified unique immune contexts of Jak3 –/– mice that cause robust IL-2-driven T cell expansion and have a clinical implication for designing a treatment strategy for human patients with loss-of-function genetic mutations of Jak3.
... Here, we addressed these questions using mouse models of ACT, where distinct subsets of donor T cells were adoptively transferred into lymphopenic host mice and then monitored for their proliferation and expansion. Specifically, we examined competition of co-transferred naïve and memory T cells during IL-7-driven lymphophenia-induced homeostatic proliferation (25)(26)(27). Interestingly, short-term adoptive transfer (1 week) resulted in a preferential expansion and accumulation of naïveorigin T cells in the LN, so that they vastly outnumbered memory-origin T cells. ...
Article
Full-text available
Naïve and memory T cells co-exist in the peripheral T cell pool, but the cellular mechanisms that maintain the balance and homeostasis of these two populations remain mostly unclear. To address this question, here, we assessed homeostatic proliferation and repopulation kinetics of adoptively transferred naïve and memory T cells in lymphopenic host mice. We identified distinct kinetics of proliferation and tissue-distribution between naïve and memory donor T cells, which resulted in the occupancy of the peripheral T cell pool by mostly naïve-origin T cells in short term (<1 week), but, in a dramatic reversal, by mostly memory-origin T cells in long term (>4 weeks). To explain this finding, we assessed utilization of the homeostatic cytokines IL-7 and IL-15 by naïve and memory T cells. We found different efficiencies of IL-7 signaling between naïve and memory T cells, where memory T cells expressed larger amounts of IL-7Rα but were significantly less potent in activation of STAT5 that is downstream of IL-7 signaling. Nonetheless, memory T cells were superior in long-term repopulation of the peripheral T cell pool, presumably, because they preferentially migrated into non-lymphoid tissues upon adoptive transfer and additionally utilized tissue IL-15 for rapid expansion. Consequently, co-utilization of IL-7 and IL-15 provides memory T cells a long-term survival advantage. We consider this mechanism important, as it permits the memory T cell population to be maintained in face of constant influx of naïve T cells to the peripheral T cell pool and under competing conditions for survival cytokines.
... In support of a role for IL-7 in naïve CD4 T cell survival, Tan et al. demonstrated a failure of transferred T cells to survive when adoptively transferred to IL-7-deficient mice (42). Also, overexpression of IL-7 was shown to enhance T cell proliferation in a lymphopenic mouse model (43). Furthermore, several studies documented enhanced peripheral T cell proliferation upon overexpression of IL-7 or the IL7R (44,45), although this was not observed in all animal models (46). ...
Article
Full-text available
There are many different pathogenic stimuli that are able to activate the immune system, ranging from microbes that include bacteria, viruses, fungi, and parasites to host-derived triggers such as autoantigens that can induce autoimmunity as well as neoantigens involved in tumorigenesis. One of the key interactions shaping immunity toward these triggers involves the encounter of antigen-processing and -presenting cells such as macrophages and dendritic cells with T cells, resulting in immune responses that are highly selective for the antigenic trigger. Research over the past few years has implicated members of the coronin protein family, in particular coronin 1, in responses against several pathogenic triggers. While coronin 1 was initially described as a host factor allowing the intracellular survival of the pathogen Mycobacterium tuberculosis, subsequent work showed it to be a crucial factor for naïve T cell homeostasis. The activity of coronin 1 in allowing the intracellular survival of pathogenic mycobacteria is relatively well characterized, involving the activation of the Ca²⁺/calcineurin pathway, while coronin 1’s role in modulating naïve T cell homeostasis remains more enigmatic. In this mini review, we discuss the knowledge on the role for coronin 1 in immune cell functioning and provide a number of potential scenarios via which coronin 1 may be able to regulate naïve T cell homeostasis.
... 2007, Т. 9, № 4-5 Характеристика иммунитета у больных раком молочной железы Считается, что в условиях лимфопении более широкая биодоступность IL-7 для циркулирующих Т-клеток, которые в норме постоянно конкурируют между собой за его связывание, приводит к запуску гомеостатической периферической экспансии (homeostatic peripheral expansion, HPE) [16,11,27]. Данный механизм направлен, в первую очередь, на быстрое количественное восстановление пула периферических Т-клеток. ...
Article
Full-text available
Some immune parameters (cell subsets in peripheral blood, spontaneous and LPS-induce production of IL-1β, IL-5, IL-7, IL-12, IL-13, IL-17, G-CSF, MCP-1 and MIP-1β in whole blood cultures) were studied in fifty patients with breast cancer (T2-3, N0-3, M0) after radiation therapy and surgical treatment. A significant decrease in lymphocytes, CD3+ and CD4+T-cell counts, CD19+B-lymphocytes, erythrocytes, hemoglobin level, and amounts of phagocytic cells was revealed. The cytokine status was characterized by prevalence of pro-inflammatory cytokines (IL-1β, IL-12, IL-17) and chemokines (MCP-1, MIP-1β), accompanied by low levels of Th2/ anti-inflammatory IL-13. Furthermore, a markedly increased production of G-CSF and IL-7 was found, thus apparently pointing to the switching of a compensatory mechanisms in response to cytoreductive effects of anti-tumor therapy. IL-7 levels in lymphopenic patients (< 1.2 х 109/L; a mean of 0.81±0.11) were significantly higher than that in an opposite group of lymphopenia-free women (> 1.2 х 109/L; a mean of 1.34±0.01). An inverse correlation (rS = –0,88; p < 0,0001) between blood lymphocyte counts and IL-7 levels allows us to suggest a mechanism of homeostatic peripheral expansion (HPE) to be involved in maintenance and restoration ofT cell homeostasis in the patients treated for breast cancer. Significance of HPE mechanism for induction of both beneficial protective tumor-specific autoimmunity and increased risk of autoimmune complications is discussed.
Article
Full-text available
We show that the lymphoid hyperplasia observed in IL-2Rα- and IL-2-deficient mice is due to the lack of a population of regulatory cells essential for CD4 T cell homeostasis. In chimeras reconstituted with bone marrow cells from IL-2Rα-deficient donors, restitution of a population of CD25+CD4+ T cells prevents the chaotic accumulation of lymphoid cells, and rescues the mice from autoimmune disease and death. The reintroduction of IL-2-producing cells in IL-2-deficient chimeras establishes a population of CD25+CD4+ T cells, and restores the peripheral lymphoid compartments to normal. The CD25+CD4+ T cells regulated selectively the number of naive CD4+ T cells transferred into T cell-deficient hosts. The CD25+CD4+/naive CD4 T cell ratio and the sequence of cell transfer determines the homeostatic plateau of CD4+ T cells. Overall, our findings demonstrate that IL-2Rα is an absolute requirement for the development of the regulatory CD25+CD4+ T cells that control peripheral CD4 T cell homeostasis, while IL-2 is required for establishing a sizeable population of these cells in the peripheral pools.
Article
Full-text available
Many antigen-specific T cells die after exposure to antigen in animals. These cells also die if they are isolated from animals shortly after activation and cultured. Various cytokines were tested for their ability to interfere with this in vitro death. Surprisingly, tumor necrosis factor α and other inflammatory cytokines did not prevent the in vitro death of activated T cells, even though these cytokines do prevent activated T cell death in animals. Therefore, the inflammatory cytokines probably act on T cells in vivo via an intermediary factor. Four cytokines, interleukin (IL)-2, IL-4, IL-7, and IL-15, did prevent activated T cell death in vitro, with IL-4 and IL-15 more effective than IL-2 or IL-7. These cytokines share a component of their receptors, the common γ chain, γc. Therefore, their collective ability to protect activated T cells from death may be mediated by signals involving γc. To assess their activity in vivo, two of the cytokines, IL-2 and IL-4, were expressed in animals at local sites of superantigen responses. Both cytokines increased the numbers of T cells found at the local sites 14 days later. Interleukin 4 was more effective than IL-2, even though IL-2 stimulates T cell proliferation better than IL-4. This result suggested that IL-4 and related cytokines can promote T cell survival in vivo as well as in vitro. The ability of these cytokines to prevent the death of activated T cells may be important at certain stages of immune responses in animals.
Conference Paper
In recent years it has become apparent that the long-term survival of T cells requires continuous contact with external stimuli. At least two types of stimuli, namely self antigens and cytokines, are involved in maintaining T cell viability. As discussed here, the factors controlling T cell survival and turnover in vivo differ considerably from one T cell subset to another.
Article
Interleukin-7, originally described as a factor controlling the survival of B-cell progenitors, has been shown by gene knock-out technology to be a non-redundant cytokine. Of all single cytokine knock-out mice, those in which the IL-7 gene has been ablated show a profound defect in lymphocyte development. Likewise, mice in which signals emanating from the corresponding receptor, whether it be by ablation of the unique alpha or common gamma chain of the receptor, or by interference with downstream signalling elements generated by this receptor complex, also show profound defects in lymphocyte differentiation. Transgenic mice over-expressing the IL-7 gene also show profound changes in lymphocyte development which, in some instances can result in the development of lymphoid tumours. Here, we review some of these aspects of IL-7 biology with particular reference to an IL-7 over-expressing transgenic mouse line in which the IL-7 transgene is controlled by the mouse MHC class II promoter.
Article
We have generated a high copy number transgenic mouse line in which expression of mouse IL-7 cDNA is under the control of the mouse MHC class II E(alpha) promoter, These mice were generated in order to see if IL-7 over-production in the thymus altered either thymocyte differentiation or the process of negative selection, Using in site hybridization, IL-7 transcripts could be detected in the thymic cortex and medulla as well as the spleen and lymph nodes of transgenic mice but was undetectable in normal controls, Phenotypic and molecular analysis of thymocytes from embryonic and adult transgenic mice failed to reveal a dramatic effect of IL-7 on thymocyte differentiation and negative selection of the TCR V-beta repertoire appeared to be intact, In peripheral lymph nodes, there was a massive (30-fold) increase in the number of T cells (CD8(+) > CD4(+)) and simultaneous presence of immature (B220(+), Ig(-)) B cells, TCR repertoire analysis showed that the expansion of peripheral T cells was polyclonal. Using the polymerase chain reaction (PCR), transgene-specific IL-7 transcripts could be detected in the thymus from day 14 of fetal development, However, using semi-quantitative PCR, there was no dramatic increase in the degree of TCR beta or TCR alpha gene rearrangements during thymocyte ontogeny in vivo, Similarly, when fetal mouse thymus lobes were cultured with IL-7 in vitro, there was no dramatic increase in the degree of TCR beta or TCR alpha gene rearrangements, We conclude that IL-7 is probably not an important differentiation factor for immature mouse thymocytes.
Article
In this special issue of The Journal of Supercomputing we have attempted to capture the most significant work that took place during the 1980s in the area of instruction-level (ILP) parallel processing. The intent is to document both the theory and the practice of ILP computing. Consequently, our emphasis is on projects that resulted in implementations of serious scope, since it is this reduction to practice that exposes the true merit and the real problems of ideas that sound good on paper.
Article
Homeostatic regulation of cell numbers is an important principle in biology. Mechanisms that function to maintain or re-establish homeostasis in the immune system include interactions among antigen-presenting cells, regulatory T cells and cytokines. The vital role that homeostatic regulation plays in maintaining a functionally intact immune system is illustrated by the perturbation of the peripheral T cell repertoire that occurs after lymphopenic incidents, which frequently provoke either exacerbated immune or autoimmune responses. Recent studies show that transient states of lymphopenia occur in viral infections and in the neonatal state and might be involved in the development of autoimmune diseases. On the positive side, lymphopenia-provoked T cell expansion might enhance weak immune responses and thereby aid the rejection of tumours or the elimination of parasites.
Article
We have generated mice that carry a germline mutation in which a large portion of the RAG-2 coding region is deleted. Homozygous mutants are viable but fail to produce mature B or T lymphocytes. Very immature lymphoid cells were present in primary lymphoid organs of mutant animals as defined by surface marker analyses and Abelson murine leukemia virus (A-MuLV) transformation assays. However, these cells did not rearrange their immunoglobulin or T cell receptor loci. Lack of V(D)J recombination activity in mutant pre-B cell lines could be restored by introduction of a functional RAG-2 expression vector. Therefore, loss of RAG-2 function in vivo results in total inability to initiate V(D)J rearrangement, leading to a novel severe combined immune deficient (SCID) phenotype. Because the SCID phenotype was the only obvious abnormality detected in RAG-2 mutant mice, RAG-2 function and V(D)J recombinase activity, per se, are not required for development of cells other than lymphocytes.