ArticlePDF Available

IL-7 Administration Alters the CD4:CD8 Ratio, Increases T Cell Numbers, and Increases T Cell Function in the Absence of Activation

Authors:

Abstract and Figures

IL-7 is vital for the development of the immune system and profoundly enhances the function of mature T cells. Chronic administration of IL-7 to mice markedly increases T cell numbers, especially CD8(+) T cells, and enhances T cell functional potential. However, the mechanism by which these effects occur remains unclear. This report demonstrates that only 2 days of IL-7 treatment is needed for maximal enhancement of T cell function, as measured by proliferation, with a 6- to 12-fold increase in the proportion of CD4(+) and CD8(+) T cells in cell cycle by 18 h of ex vivo stimulation. Moreover, a 2-day administration of IL-7 in vivo increases basal proliferation by 4- and 14-fold in CD4(+) and CD8(+) T cells, respectively. These effects occur in the absence of cytokine production, increases in most activation markers, and changes in memory markers. This enhanced basal proliferation is the basis for the increase in T cell numbers in that IL-7 induces an additional 60% and 85% of resting CD4(+) and CD8(+) T cells, respectively, to enter cell cycle in mice given IL-7 for 7 days. These results demonstrate that in vivo administration of IL-7 increases T cell numbers and functional potential via a homeostatic, nonactivating process. These findings may suggest a unique clinical niche for IL-7 in that IL-7 therapy may increase T cell numbers and enhance responses to specific antigenic targets while avoiding a general, nonspecific activation of the T cell population.
Content may be subject to copyright.
IL-7 Administration Alters the CD4:CD8 Ratio, Increases T
Cell Numbers, and Increases T Cell Function in the Absence of
Activation
1
Lisa A. Geiselhart,* Courtney A. Humphries,* Theresa A. Gregorio,
Sherry Mou,
Jeffrey Subleski,* and Kristin L. Komschlies
2†
IL-7 is vital for the development of the immune system and profoundly enhances the function of mature T cells. Chronic admin-
istration of IL-7 to mice markedly increases T cell numbers, especially CD8
T cells, and enhances T cell functional potential.
However, the mechanism by which these effects occur remains unclear. This report demonstrates that only 2 days of IL-7
treatment is needed for maximal enhancement of T cell function, as measured by proliferation, with a 6- to 12-fold increase in the
proportion of CD4
and CD8
T cells in cell cycle by 18 h of ex vivo stimulation. Moreover, a 2-day administration of IL-7 in vivo
increases basal proliferation by 4- and 14-fold in CD4
and CD8
T cells, respectively. These effects occur in the absence of
cytokine production, increases in most activation markers, and changes in memory markers. This enhanced basal proliferation is
the basis for the increase in T cell numbers in that IL-7 induces an additional 60% and 85% of resting CD4
and CD8
T cells,
respectively, to enter cell cycle in mice given IL-7 for 7 days. These results demonstrate that in vivo administration of IL-7
increases T cell numbers and functional potential via a homeostatic, nonactivating process. These findings may suggest a unique
clinical niche for IL-7 in that IL-7 therapy may increase T cell numbers and enhance responses to specific antigenic targets while
avoiding a general, nonspecific activation of the T cell population. The Journal of Immunology, 2001, 166: 3019–3027.
I
nterleukin 7 is a 25-kDa glycoprotein produced by thymic
and intestinal epithelial cells, bone marrow stromal elements
and keratinocytes and has been shown to be an essential
growth factor for B and T lineage cells (reviewed in Ref. 1). IL-7
also acts as a T cell costimulus and can enhance in vitro T cell
responses in an Ag-specific fashion when added simultaneously
with various stimuli (2–7). In vitro, IL-7 in the absence of any
other stimulus has been shown to induce proliferation of fresh T
cells in a dose-dependent fashion in some reports (3, 4, 8) but not
in others (5, 9). In addition, reports indicate that human T cells
may proliferate more robustly to IL-7 than mouse T cells, and
some results suggest that the proliferation induced by IL-7 is de-
pendent on the presence of APC (2–4, 8, 9). Although IL-7 does
not appear to switch T cells from CD45RA to CD45RO (10), up-
regulation of activation markers such as CD25, CD98, CD71,
CD11a, and CD40 ligand has been reported in vitro (3, 8, 11). In
addition, less mature T cells (such as those from human cord
blood) appear to proliferate more vigorously to IL-7 than do T
cells from adults (12).
In vivo administration of IL-7 results in the increase of B lin-
eage cell and T cell numbers with a preferential increase in CD8
T cells (13–15). Although this increase in T cell numbers appears
to be predominantly a thymic independent event (15), the mech-
anism by which the increase in T cell numbers occurs remains to
be determined. In addition, T cells from IL-7-treated mice generate
enhanced CTL and proliferative responses to subsequent ex vivo
stimulation. However, it has not been determined whether the in-
creased functional ability of T cells from IL-7-treated mice is due
to a direct alteration in the biological status of individual T cells or
whether it is a result of the alteration in the CD4:CD8 subset ratio
due to the disproportionate increase in CD8
T cells that simul-
taneously occurs.
The results presented in this report demonstrate that T cells from
IL-7-treated mice acquire enhanced functional capacity, as mea-
sured by proliferation, before the disproportionate increase in
CD8
T cells and resultant CD4:CD8 ratio alteration and involves
the enhanced activity of both CD4
and CD8
T cells. The en-
hanced functional capacity appears to be attributable to the ability
of IL-7 to increase the level of basal proliferation. Thus, T cells
from IL-7-treated mice already have the cell cycle machinery in
place to respond in an enhanced fashion to a subsequent stimula-
tion. This IL-7-induced proliferation appears to be nonactivating in
that these T cells are not producing cytokine. Moreover, adminis-
tration of IL-7 in vivo does not induce alterations in most activa-
tion and memory markers examined. This is in contrast to in vitro
models that have shown an up-regulation of several activation
molecules (3, 8, 11), thereby demonstrating that in this regard,
previously reported in vitro results are not representative of what
occurs in vivo. Finally, our results demonstrate that the increase in
T cell numbers after IL-7 administration is attributable at least in
part to the ability of IL-7 to induce additional T cells to enter cell
*Laboratory of Experimental Immunology, Division of Basic Sciences, and
Intra-
mural Research Support Program, Science Applications International Corp. Frederick,
National Cancer Institute-Frederick Cancer Research and Development Center, Fred-
erick, MD 21702
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.
Received for publication August 7, 2000. Accepted for publication December 18,
2000.
1
The content of this publication does not necessarily reflect the views or policies of
the Department of Health and Human Services, nor does mention of trade names,
commercial products, or organization imply endorsement by the U.S. Government.
This project has been funded in whole or in part with Federal funds from the National
Cancer Institute, National Institutes of Health, under Contract No. NO1-CO-56000.
Animal care was provided in accordance with the procedures outlined in Guide for the
Care and Use of Laboratory Animals (National Institutes of Health Publication No.
86-23, 1985).
2
Address correspondence and reprint requests to Dr. Kristin L. Komschlies, Intra-
mural Research Support Program, Science Applications International Corp. Frederick,
National Cancer Institute-Frederick Cancer Research and Development Center, Build-
ing 560, Room 31-93, Frederick, MD 21702-1201. E-mail address: komschliesk@
mail.ncifcrf.gov
Copyright © 2001 by The American Association of Immunologists 0022-1767/01/$02.00
cycle. These results are important for the potential use of IL-7
clinically in that IL-7 increases T cell numbers and functional ca-
pacity via a nonactivating process. Thus, polyclonal activation of
T cells does not occur with IL-7 treatment in vivo and only T cells
with specificity to a particular Ag may respond with enhanced
vigor to such antigenic stimuli as unique tumor Ags or DNA vac-
cines encoding these Ags.
Materials and Methods
Mice
C57BL/6 and C57BL/6-CD45.1 mice were used at 2–3 mo of age and were
obtained from the Animal Production Area of the National Cancer Insti-
tute-Frederick Cancer Research and Development Center (Frederick, MD).
Breeding pairs of C57BL/6 (CD45.2)-IL-7R knockout mice were pur-
chased from The Jackson Laboratory (Bar Harbor, ME) and bred in our
animal facility to provide mice for this study. All mice were maintained
under specific pathogen-free conditions.
Treatment of mice
IL-7 (recombinant human IL-7) was generously provided by Sanofi Re-
cherche (Gentilly, France). The IL-7 had a biologic activity of 5.4 10
7
U/mg, as measured by the proliferation of a murine pre-B cell line (16); the
endotoxin levels were 1.3 EU/mg of IL-7. Mice were injected i.p. twice
a day for a varying number of days with HBSS (without Ca
2
,Mg
2
,or
phenol red; BioWhittaker, Walkersville, MD) plus 0.1% normal mouse
serum (NMS)
3
as a vehicle control or with IL-7 at 10
g/injection diluted
in HBSS plus 0.1% NMS at 0.2 ml/injection. Mice were euthanized the day
after treatment completion, and their tissues were then analyzed.
Fluorescent dye labeling of cells and injection into recipient
mice
In one set of experiments a single-cell suspension of peripheral lymph node
(inguinal, axillary, and brachial) cells prepared as previously described
(15) from C57BL/6-CD45.1 mice were labeled with CFSE (Molecular
Probes, Eugene, OR) to monitor proliferation (17, 18). Cells were resus-
pended in PBS at 20 10
6
cells/ml. One milliliter of 200 nM CFSE (in
PBS) was added per 1 ml of cell suspension, followed by mixing and
incubation at room temperature for 15 min in the dark. After the incubation
period, 1 ml of FCS per 1 ml of CFSE-cell suspension was added to in-
activate the labeling reaction. Cells were washed once in PBS and counted.
CFSE-labeled cells were injected i.v. into C57BL/6-(CD45.2)-IL-7R
knockout mice. Twenty-four hours after injection of the cells, mice were
treated with HBSS plus 0.1% NMS or IL-7 as indicated above.
T cell stimulation assay
Peripheral lymph node cells were cultured as described previously (15) at
a concentration of 2 10
5
cells per well with medium alone or a 1:10
supernatant of anti-CD3 generated in vitro by the hybridoma clone 145/
2C11 (19) and 0.5
g/ml of anti-CD28 (BD PharMingen, San Diego, CA).
Cell cultures were pulsed with 0.5
Ci of [
3
H]thymidine (Amersham Life
Science, Piscataway, NJ) at the initiation of culture. After culture, the cells
were recovered by using a Harvester 96 (Tomtec, Hamden, CT). Prolifer-
ation was assessed in triplicate by measuring the amount of cellular incor-
poration of [
3
H]thymidine in cpm with a 1450 MicroBeta TRILUX liquid
scintillation and luminescence counter (Wallac, Turku, Finland). Cells
(2 10
6
/ml) also were stimulated in vitro with PMA (20 ng/ml) and
ionomycin (1
g/ml) or Con A (5
g/ml).
Surface phenotyping of cells
Single-cell suspensions of peripheral lymph nodes or spleens were pre-
pared in HBSS plus 0.5% BSA. RBC were lysed using ACK lysing buffer
(BioWhittaker). Cells were labeled with optimally titered Abs, and 10
4
cells were analyzed for the percentage of cells bearing a particular mark-
er(s) by using a FACScan flow cytometer affixed with a doublet discrim-
ination module (Becton Dickinson, Mountain View, CA) as described pre-
viously (20). Analysis was performed using CellQuest software (Becton
Dickinson). Subset analysis in Fig. 1 was performed using PE-conjugated
anti-CD4 mAb (Becton Dickinson) and biotin-conjugated anti-CD8 mAb
(Becton Dickinson) developed with streptavidin-RED670 (Life Technolo-
gies, Gaithersburg, MD). Surface expression of activation markers on T
cell subsets was determined by using FITC-conjugated mAb to CD25,
CD69, and CD71 (BD PharMingen) individually combined with PE-con-
jugated anti-CD4 mAb and biotin-conjugated anti-CD8 developed with
streptavidin-RED670. In addition, PE-conjugated mAb to CD137 (BD
PharMingen) was individually combined with FITC-conjugated anti-CD4
mAb (clone H129.19; Ref. 21; conjugated in our laboratory) or FITC-
conjugated anti-CD8 mAb (BD PharMingen) was used. Surface expression
of memory markers on T cell subsets were determined by using FITC-
conjugated mAb to CD44 and PE-conjugated mAb to CD62L (BD PharM-
ingen) combined with biotin-conjugated mAb to CD4 (BD PharMingen) or
biotin-conjugated mAb to CD8 developed with streptavidin-RED670.
Where required, cells from C57BL/6-CD45.1 mice (donor-origin) and
C57BL/6-CD45.2-IL-7R
/
mice (host-origin) were detected by using anti-
CD45.1 mAb (clone A-20-1.7; Ref. 22) or anti-CD45.2 mAb (clone
104.2.1; Ref. 22), respectively, developed with a PE-conjugated goat anti-
mouse IgG2a specific polyclonal antiserum (Southern Biotechnology As-
sociates, Birmingham, AL). In this system, CD4
and CD8
T cells were
detected by using biotin-conjugated anti-CD4 (clone H129.19, conjugated
in our laboratory) or anti-CD8 mAb developed with streptavidin-RED670.
Cell cycle analysis
Leukocytes were labeled with FITC-conjugated anti-CD4 or anti-CD8 (BD
PharMingen) to distinguish their cell surface phenotype as described
above. Surface-labeled cells were then permeabilized by resuspension in
saponin buffer (0.1% BSA, 0.01 M HEPES, 0.1% saponin in PBS) at a
concentration of 0.5 10
6
cells/ml followed by centrifugation at 1500 rpm
for 5 min at 4°C. The supernatant was decanted and the pellet was resus-
pended in 0.5 ml/10
6
cells of saponin buffer containing 200
g/ml of pro
-
pidium iodide (Sigma, St. Louis, MO) and 50
g/ml of RNase (Puregene,
Minneapolis, MN) followed by incubation for 15 min at 4°C. Labeled cells
were analyzed by using a FACScan flow cytometer affixed with a doublet
discrimination module to include only single cells in the cell cycle analysis.
Data were analyzed using CellQuest software.
Analysis of 5-bromo-2-deoxyurindine (BrdU) incorporation by
lymph node cells
Leukocytes were cultured at 10
6
cells/ml in medium alone or anti-CD3 and
anti-CD28 as indicated above. For each culture condition, cells were cul-
tured either with or without 10
g/ml of BrdU (Sigma). After culture, cells
were washed in HBSS. Cell surface labeling was performed as above in
PBS without azide by using PE-conjugated anti-CD4 mAb and biotin-
conjugated anti-CD8 mAb developed with streptavidin-RED670 to distin-
guish T cell subsets at 10
6
cells per sample in a 96-well round-bottom plate.
After the final wash, the supernatant was removed and the pellets were
resuspended in 0.1 ml of PBS and transferred to 12 75-mm polystyrene
tubes containing 1 ml of PBS followed by centrifugation at 1500 rpm for
5 min at 4°C. The supernatant was removed and 0.5 ml of cold 0.15 M
NaCl was added per sample. While gently vortexing, 1.2 ml cold 95%
ethanol was added dropwise to each sample followed by a 30-min incu-
bation on ice. After incubation, 2 ml of PBS was added to the samples
followed by centrifugation at 1800 rpm for 5 min at 4°C. The supernatant
was removed and the cells were permeabilized by slowly vortexing and
adding 1 ml of PBS containing 1% paraformaldehyde (Sigma) and 0.01%
Tween 20 (Sigma). Cells were incubated for 30 min at room temperature
followed by centrifugation at 1800 rpm for 6 min at 4°C. Each pellet was
resuspended slowly while vortexing in 1 ml of DNase I (DNase I from
bovine pancreas at 50 Kunitz U/ml in 4.2 mM MgCl/0.15 M NaCl, pH 5;
Roche Molecular Biochemicals, Indianapolis, IN). Cells were incubated for
10 min at room temperature then washed in 2 ml PBS followed by cen-
trifugation at 1800 rpm for 6 min at 4°C. Twenty microliters of optimally
titered FITC-conjugated anti-BrdU mAb (BD PharMingen) diluted in PBS
was added; the cells were gently mixed, incubated for 30 min at room
temperature, and washed in 2 ml of PBS followed by centrifugation at 1800
rpm for 6 min at 4°C. Samples were resuspended in 0.2 ml of PBS and
placed on ice in the dark until analyzed by using a FACScan flow cytom-
eter. Cells cultured without BrdU were used as nonspecific binding controls
for the anti-BrdU mAb.
Immune complex kinase assay
Lymph node cells were washed twice in HBSS and disrupted in 1 ml of
lysis buffer (1% (v/v) Triton X-100, 50 mM NaCl, 10 mM Tris-HCl (pH
7.5), 5 mM EDTA, 30 mM sodium pyrophosphate, 5 mM NaF, 25 mM
-glycerolphosphate, 5 mM sodium orthovanadate, 0.1% p-nitrophe-
nylphosphate, 1 mM PMSF) at 4 10
7
cells/ml. Cell lysates were clarified
by centrifugation (5000 g for 20 min at 4°C), and the protein concen-
tration was determined with the bicinchoninic acid protein detection kit
3
Abbreviations used in this paper: NMS, normal mouse serum; BrdU, 5-bromo-2-
deoxyurindine; Rb, retinoblastoma.
3020 IL-7 INCREASES T CELL NUMBERS AND FUNCTION WITHOUT ACTIVATION
(Pierce, Rockford, IL). For Cdk2 kinase assay, 500
g of protein from
clarified cell lysates were incubated with 1.5
g of anti-Cdk2 (Santa Cruz
Biotechnology, Santa Cruz, CA); after 3 h, 50
l of a 1:1 slurry of protein
G-agarose was added and incubated for an additional 60 min. The immune
complexes were then washed three times with lysis buffer and two times in
a buffer of 10 mM HEPES, pH 7.2. The immune complexes were resus-
pended in 50
l of kinase buffer (10 mM HEPES, pH 7.2, 10 mM MgCl
2
,
10 mM MnCl
2
,50
Ci/ml [
-
32
P]ATP, and 3
g of histone H1, a substrate
for cyclin-dependent kinases). The reaction was terminated after 20 min at
room temperature by the addition of 3 SDS sample buffer, and the kinase
mixture was separated through a 10% polyacrylamide SDS gel and trans-
ferred to an Immobilon-P membrane (Millipore, Bedford, MA). Autora-
diography was performed to detect protein radiolabeled in the immune
complex kinase assay.
Immunoblotting
For detection of cyclin E and retinoblastoma (Rb) protein, lymph node cells
were solubilized in 1 ml of solubilization buffer (50 mM HEPES, pH 7.4;
15 mM EGTA; 137 mM NaCl; 15 mM MgCl
2
; 0.1% Triton X-100; 10 mM
-glycerophosphate, 1 mM Na
3
VO
4
; 1 mM PMSF; and 1
g/ml aprotinin/
leupeptin). Insoluble material was removed by centrifugation (5000 g for
20 min at 4°C), and 20
g of total protein was resolved by 12% SDS-
PAGE, and transferred to an Immobilon-P membrane. The membrane was
blocked in TBST (20 mM Tris, pH 7.6, 137 mM NaCl, and 0.1% Tween
20) containing 5% nonfat dry milk (6 h), washed twice, and then incubated
overnight with specific anti-Cdk2 antiserum, anti-cyclin E antiserum (Santa
Cruz Biotechnology), or anti-Rb mAb (clone G3-245; BD PharMingen).
After vigorous washing, blots were incubated first with a biotinylated sec-
ondary Ab (anti-rabbit or mouse, as appropriate), then with peroxidase-
conjugated streptavidin, and developed by ECL.
Results
Enhanced T cell response requires only 2 days of IL-7 treatment
in vivo
Previous studies have shown that chronic administration of IL-7 to
mice or expression of an IL-7 transgene results in an increase in T
cell numbers (13–15, 23, 24). Other studies revealed an ability of
IL-7 to prime mature T cells for polyclonal activation stimuli (e.g,
anti-CD3) (15). However, little is known about whether the ability
of IL-7 to prime T cells for activation is causally linked to the
induction of proliferation. To address this issue, mice were treated
twice a day with IL-7 (10
g/injection) for 2, 4, or 7 days. After
treatment, lymph node cells were stimulated in culture with anti-
CD3 and anti-CD28 for 18 h, and proliferation was monitored. The
results in Fig. 1A demonstrate that IL-7 administration for 2, 4, or
7 days resulted in increased proliferation by 5.0-, 5.1-, or 6.1-fold,
respectively, over that of vehicle control (HBSS plus 0.1% NMS).
Thus, IL-7 treatment induces peak priming for subsequent en-
hancement of proliferative response by 2 days. Moreover, as
shown in Fig. 1B, this enhanced response occurs independently of
the disproportionate increase in CD8
cells induced by IL-7 treat
-
ment after 4 days or more of IL-7 administration.
In a similar experiment with 2 days of IL-7 administration, cell
cycle analysis was performed to specifically identify the respond-
ing cell type(s) 18 h after the initiation of ex vivo stimulation with
anti-CD3 and anti-CD28. The results demonstrate that 1.96% and
3.60% of CD4
and CD8
T cells, respectively, were in S phase
or G
2
/M in the vehicle control-treated mice (Fig. 2
). However, in
lymph node cell cultures from mice treated with IL-7, 11.43% and
42.56% of CD4
and CD8
T cells, respectively, were in cycle.
This represents a 5.8-fold increase in CD4
T cells and an 11.8-
fold increase in CD8
T cells that were in cycle at 18 h of culture
with anti-CD3 and anti-CD28 when cells from IL-7-treated mice
were compared with controls. In addition, lymph node cells from
2-day HBSS- or IL-7-treated mice were cultured ex vivo for 24 h
with anti-CD3 and anti-CD28 in the presence of BrdU to deter-
mine the proportion of cells that entered cell cycle over the 24-h
period. The results in Fig. 3 (upper panels) demonstrate that the
total lymph node cells from IL-7-treated mice have a dramatic
increase in the proportion of cells entering cell cycle compared
with cells from control mice whose levels were only slightly above
background levels. More specifically, Fig. 3 (middle and lower
panels) shows that, after subtraction of background values, 1.4%
of CD4
T cells and 4.9% of CD8
T cells from vehicle control
mice had entered cell cycle. In contrast, 17.0% of CD4
T cells
and 41.4% of CD8
T cells from IL-7-treated mice had entered
cell cycle. Thus, pretreatment with IL-7 in vivo results in an 12-
and 8-fold increase in the number of CD4
and CD8
T cells,
respectively, that enter cell cycle within the first 24 h of ex vivo
stimulation compared with cells from vehicle control mice.
IL-7 treatment in vivo results in an increase in the basal level of
T cell proliferation
One possible explanation for the accelerated proliferative capacity
of lymph node T cells from IL-7-treated mice is that IL-7 induces
T cells to move at least partially into cell cycle. The results in Fig.
1 support this possibility in that lymph node cells from mice
treated with IL-7 for 2, 4, or 7 days have a 7.5-, 9.0-, or 11.1-fold
FIGURE 1. Two days of in vivo administration of IL-7 enhances sub-
sequent T cell responses independently of the disproportionate increase in
CD8
T cells. C57BL/6 mice were injected i.p. twice a day with HBSS
plus 0.1% NMS (HBSS; vehicle control) for 7 days or 10
g/injection of
IL-7 for 2, 4, or 7 days. After cessation of treatment, single-cell suspen-
sions were prepared from pooled peripheral lymph nodes from 6–15 mice/
group. A, Cells were cultured in medium alone or with anti-CD3 mAb and
anti-CD28 mAb for 18 h. [
3
H]Thymidine was added at the initiation of
culture to assess the proliferative response. Each bar represents the mean
cpm of three replicates SD. B, Cells were stained with anti-CD4 or
anti-CD8 mAb. Cell surface expression of these phenotypic markers was
determined by flow cytometric analysis. The percentage of positive cells
bearing a particular marker was multiplied by the mean of the total number
of lymph node leukocytes per mouse (six lymph nodes per mouse) to
determine the number of cells within the subset.
3021The Journal of Immunology
increase, respectively, in [
3
H]thymidine incorporation when cul
-
tured for 18 h in medium alone compared with HBSS control
lymph node cells.
Biochemical analysis of lymph node cells from mice treated
with IL-7 for 2 days revealed an increased level of Cdk2 kinase
activity as evidenced by the presence of phosphorylated histone
H1, a substrate of Cdk2 kinase; whereas HBSS-treated control
lymph node cells had no detectable level of Cdk2 kinase activity
(Fig. 4A, upper panel). This increase was not due to alterations in
the total amount of Cdk2 kinase, as the level of Cdk2 kinase was
equivalent in the two groups (Fig. 4A, lower panel). Moreover,
whole-cell lysates from lymph node cells from IL-7-treated mice
had elevated levels of cyclin E (Fig. 4B) and phosphorylated Rb
compared with cells from mice that had not received IL-7 (Fig.
4C). These results demonstrate that IL-7 administration can in-
crease activity/levels of these components critical for the move-
ment of cells from G
0
through G
1
and toward the S phase of cell
cycle.
To determine whether IL-7 had an effect on the basal prolifer-
ation levels of T cell subsets, cell cycle analysis was performed as
described above with lymph node cells from mice treated with
IL-7 or HBSS (vehicle control) for 2 days. The results in Fig. 5
demonstrate that in the vehicle control group, 0.88% of the CD4
T cells and 0.70% of the CD8
T cells were in either S phase or
G
2
/M. In contrast, in the IL-7-treated group, 3.77% of the CD4
T cells and 10.21% of the CD8
T cells were in either S phase or
G
2
/M. Thus, a 2-day administration of IL-7 in vivo induces a 4.3-
and 14.6-fold increase in CD4
and CD8
T cells, respectively,
that are in S/G
2
/M. Furthermore, in vivo IL-7 treatment results in
a differential response by T cell subsets in that 3-fold more CD8
T cells are in S/G
2
/M at a given time point compared with CD4
T cells. In contrast, cells from control-treated mice have an ap-
proximately equal proportion of CD4
and CD8
T cells in
S/G
2
/M. These observations are further supported when lymph
node cells from 2-day HBSS- or IL-7-treated mice were cultured
ex vivo for 24 h in the presence of BrdU. The results in Fig. 6
(upper panels) show that over the 24-h culture period there was no
detectable level of BrdU incorporated above background values in
total lymph node cells from control-treated mice. However, lymph
node cells from IL-7-treated mice had detectable levels of BrdU
incorporation. Detailed examination revealed that after subtraction
of background, 4.0% of the total CD4
T cells and 10.7% of the
total CD8
T cells from IL-7-treated mice entered cell cycle.
These results are similar to that of the cell cycle data and clearly
demonstrate that in vivo administration of IL-7 increases the basal
proliferation level in both T cell subsets, but preferentially in
CD8
T cells.
The enhanced basal proliferation induced by in vivo IL-7
administration does not result in a concomitant induction of
most activation markers, cytokine production, or alterations in
memory markers
To determine the effect of IL-7 on the activation and the naive/
memory status of T cells, lymph node cells from mice given a
2-day treatment of HBSS plus 0.1% NMS or IL-7 were analyzed
by using flow cytometry to determine the surface expression of
activation and memory markers on T cell subsets. The results in
Fig. 7 show that 2 days of IL-7 treatment in vivo (dark solid line)
does not change the cell surface expression of the activation mark-
ers CD25, CD69, and CD137 or the memory markers CD44 and
CD62L on CD4
or CD8
T cells compared with cells from
HBSS control-treated mice (dashed line). However, CD71 is up-
regulated but only on the CD8
T cells from IL-7-treated mice.
Although IL-7 induces the expression of CD71 on CD8
T cells,
the intensity of expression is less than that induced on stimulation
with PMA and ionomycin (light solid line). Based upon these
markers, IL-7 treatment in vivo does not have an overall activating
effect on T cells in that most of the activation markers remained
unchanged. Furthermore, the naive/memory status of T cells is not
altered with 2 days of IL-7 treatment. However, IL-7 has a limited
and differential effect on CD8
T cell activation compared with
CD4
T cells based upon the up-regulation of CD71.
To determine whether IL-7 treatment could induce another T cell
function in addition to proliferation, the ability of cells from IL-7-
treated mice to produce cytokine was examined. Lymph node cells
from mice treated with HBSS or IL-7 for 2 days were cultured for
FIGURE 2. IL-7 administration increases the propor-
tion of T cells that are in S/G
2
/M of the cell cycle at a
given time point in response to subsequent stimulation.
C57BL/6 mice were injected i.p. twice a day for 2 days
with HBSS plus 0.1% NMS (control) or IL-7 (10
g/
injection). Peripheral lymph node cells then were stim-
ulated in vitro with mAb to CD3 and CD28 for 18 h.
After culture, cells were surface-labeled with fluoro-
chrome-conjugated mAb to CD4 and CD8 to discrimi-
nate the two T cell subsets followed by treatment with
propidium iodide for detection of cell-cycle status by
using flow cytometric analysis. The histograms were
generated by gating on either the CD4
or CD8
T cell
subset and displaying the cell cycle status for that par-
ticular subset.
3022 IL-7 INCREASES T CELL NUMBERS AND FUNCTION WITHOUT ACTIVATION
24 h in medium. After culture, supernatants were assayed by ELISA
to determine whether the cytokines IL-2, IL-4, GM-CSF, or IFN-
were produced. No detectable levels of any of the cytokines examined
were found in the supernatants from either group. In contrast, super-
natants from control cells stimulated with anti-CD3 and anti-CD28
contained considerable amounts of each of these cytokines (data not
shown). Thus, while in vivo administration of IL-7 increases basal
proliferation levels in T cells, this is not an activation-induced prolif-
eration as it is not associated with other hallmarks of cell activation
including cytokine production and up-regulation of the cell surface
markers typically associated with T cell activation.
Increase in T cell numbers induced by IL-7 administration in
vivo is due, at least in part, to proliferation of peripheral T cells
The ability of in vivo administration of IL-7 to induce increased
basal proliferation suggested that the IL-7-induced increase in T
cell numbers may occur via proliferation of peripheral T cells. To
test this hypothesis, lymph node cells from C57BL/6-CD45.1 con-
genic mice were labeled ex vivo with CFSE, a fluorescent dye that
binds irreversibly to cellular components. On division, CFSE is
distributed evenly between daughter cells and the mean fluores-
cence halves accordingly. CFSE labeled cells were injected i.v.
into C57BL/6-CD45.2-IL-7 receptor knockout mice. After allow-
ing the cells 24 h to home to lymphoid tissues, mice were injected
i.p. twice a day for 7 days with HBSS or IL-7. After treatment, the
splenocytes were examined by using flow cytometric analysis to
enumerate the number of donor-origin (CD45.1
) T cells and to
determine whether they had undergone proliferation. Fig. 8 dem-
onstrates that 71.5% of the donor-origin CD4
T cells from a
representative mouse treated with HBSS were in the nonprolifer-
ating portion (highest intensity of CFSE), whereas only 13.7% of
the donor-origin CD4
T cells from IL-7-treated mice had an
equivalent level of CFSE intensity. Similarly, 71.4% of the donor-
origin CD8
T cells from vehicle control-treated mice fell in this
range of CFSE intensity; in contrast, only 2.3% of the donor-origin
CD8
T cells from IL-7-treated mice fell into this range (Fig. 8).
FIGURE 3. IL-7 administration induces enhanced T cell proliferation in
response to subsequent stimulation. C57BL/6 mice were injected i.p. twice
a day for 2 days with HBSS plus 0.1% NMS (control) or IL-7 (10
g/
injection). Peripheral lymph node cells were stimulated in culture with
mAb to CD3 and CD28 for 24 h. At initiation of culture, BrdU was added
to determine the number of cells that entered cell cycle. After culture, cells
were surface-labeled with fluorochrome-conjugated mAb to CD4 and CD8,
then fixed, permeablized, and labeled intracellularly with a fluorochrome-
conjugated mAb to BrdU for flow cytometric analysis. The data repre-
sented in the plots are the profiles of the total leukocyte population. In the
upper panels, the solid line indicates the proportion of cells that incorpo-
rated BrdU and the dashed line indicates the nonspecific background bind-
ing of the anti-BrdU mAb that was generated using cells cultured under
similar conditions but in the absence of BrdU. The percentage of nonspe-
cific binding was 0.3% per quadrant.
FIGURE 4. IL-7 treatment induces cell cycle proteins and kinase activ-
ity. C57BL/6 mice were injected i.p. twice a day for 2 days with HBSS plus
0.1% NMS or IL-7 (10
g/injection). Peripheral lymph node cells were
pooled by treatment group. A, Top, An immune complex kinase assay was
performed with Cdk2 immunoprecipitated (IP) from equivalent protein
amounts. Histone H1 was included as a phosphorylation substrate for
Cdk2. Samples were resolved with SDS-PAGE, transferred to an Immo-
bilon-P membrane, and radiolabeled proteins were detected with autora-
diography. Bottom, Immunoblotting (IB) with Cdk2-specific antiserum, il-
lustrates the total amount of Cdk2 protein (33-kDa doublet) in each sample.
B and C, Levels of cyclin E (50 kDa; B) or phosphorylated Rb protein
(105–110 kDa; C) were determined by using whole-cell lysates. An equiv-
alent amount of protein was loaded in each sample, and the samples were
resolved with SDS-PAGE and transferred to Immobilon-P membrane. Im-
munoblotting was performed with antiserum specific for cyclin E or Rb,
respectively. Hyperphosphorylated Rb (pRb) migrates more slowly than
the less phosphorylated forms lower in the blot. In C, the top band in all
lanes is attributable to nonspecific binding, and the control is actively pro-
liferating MOLT-4 cells.
3023The Journal of Immunology
Therefore, there is a 4- and 16.7-fold increase in the proportion of
CD4
and CD8
T cells, respectively, undergoing proliferation
compared with T cells from mice treated with HBSS. To determine
whether IL-7 induced additional cells to proliferate, the number of
cells that the fell into the nonproliferating vs proliferating catego-
ries were calculated in Fig. 9. The number of CD4
and CD8
T
cells that remained in a nonproliferation status was decreased by
59.7% and 84.6%, respectively, in the spleens from mice treated
with IL-7. Furthermore, these data demonstrate for the first time
that IL-7 acts directly on T cells (i.e., IL-7R
/
donor-origin T
cells) to achieve these effects as the recipient cells (IL-7R
/
)in
the microenvironment are genotypically unable to respond to IL-7.
In addition, the increase in donor-origin T cell numbers is not due
to differentiation of T cell precursors that are absent in lymph node
cell inoculum. Moreover, these data demonstrate that IL-7 admin-
istration preferentially induces CD8
T cells to proliferate. Thus,
IL-7 increases T cell numbers in vivo at least in part by directly
inducing additional nonprecursor peripheral T cells to undergo
proliferation.
Discussion
Although in vivo administration of IL-7 has been demonstrated to
enhance T cell functional capacity (15) and increase T cell num-
bers, particularly CD8
T cells (13–15), little is known regarding
the kinetics, responsible T cell subset(s), and mechanism(s) by
which these in vivo phenomena occur. To initiate investigation of
these issues, we speculated that the enhanced T cell functional
capacity, which we demonstrated in a previous report where IL-7
was administered in vivo for 7 days (15), may result from a change
in the composition of the T cell population from an approximately
equal proportion of CD4
and CD8
T cells in controls to a ma
-
jority of CD8
cells in the IL-7-treated group. In this report we
have demonstrated that the enhanced T cell functional capacity
induced by in vivo administration of IL-7 occurs after as little as
2 days of treatment with IL-7 (Fig. 1A) and before the alteration in
the CD4:CD8 ratio (Fig. 1B) and the increase in pre-B cell num-
bers (data not shown) that occurs after 4 or more days of IL-7
treatment. In addition, in vivo IL-7 administration enhances the
proliferative response to a subsequent ex vivo stimulus of the
CD4
and CD8
T cell subsets by 6- and 12-fold, respectively
FIGURE 6. In vivo treatment with IL-7 induces T cell proliferation.
C57BL/6 mice were injected i.p. twice a day for 2 days with HBSS plus
0.1% NMS (control) or IL-7 (10
g/injection). Peripheral lymph node cells
were cultured for 24 h in medium alone with BrdU. After culture, cells
were surface-labeled with fluorochrome-conjugated mAb to CD4 and CD8,
then fixed, permeabilized, and labeled intracellularly with a fluorochrome-
conjugated mAb to BrdU for flow cytometric analysis. The data repre-
sented in the plots are the profiles of the total leukocyte population. In the
upper panels, the solid line indicates the proportion of cells that incorpo-
rated BrdU, and the dashed line indicates the nonspecific binding control as
described in Fig. 3. The percentage of nonspecific binding was 0.5% per
quadrant.
FIGURE 5. IL- 7 administration increases the propor-
tion of T cells that are in cell cycle. C57BL/6 mice were
injected i.p. twice a day for 2 days with HBSS plus 0.1%
NMS (control) or IL-7 (10
g/injection). Peripheral
lymph node cells from these mice were surface-labeled
with fluorochrome-conjugated mAb to CD4 and CD8,
then treated with propidium iodide for detection of cell-
cycle status by using flow cytometric analysis. The his-
tograms were generated by gating on either the CD4
or
CD8
T cell subset and displaying the cell cycle status
for that particular subset.
3024 IL-7 INCREASES T CELL NUMBERS AND FUNCTION WITHOUT ACTIVATION
(Fig. 2). Furthermore, the ability of in vivo administration of IL-7
to enhance T cell function is not merely due to the reported co-
stimulatory effects of IL-7 (2–5) as a result of carry-over of IL-7
from the in vivo treatment into the ex vivo culture. This is evi-
denced by the fact that addition of IL-7 in culture failed to enhance
the proliferative response of lymph node cells from HBSS control-
treated mice stimulated with anti-CD3 and anti-CD28 by 24 h of
culture (data not shown). Thus, the enhancing effect of IL-7 on T
cell function, as measured by proliferation, occurs in both T cell
subsets and is independent of the disproportionate increase in
CD8
T cells. In terms of the clinical use of IL-7, these results
demonstrate that the functional enhancing properties of IL-7 may
be separated from the ability of IL-7 to increase cell numbers by
varying the length of treatment. Thus, only short periods of IL-7
treatment may be needed to enhance T cell function in patients.
In addition to the ability of IL-7 administration in vivo to en-
hance the proliferative response of T cells to a subsequent ex vivo
stimulus, results presented here demonstrate that IL-7 administra-
tion for as little as 2 days results in increased basal proliferation of
T cells. This increase in basal proliferation may be the basis for the
enhanced proliferative response to subsequent stimulation induced
FIGURE 9. In vivo administration of IL-7 acts directly to induce IL-
7R
donor T cells to enter cell cycle in IL-7R
/
recipient mice. The
results from Fig. 8 were analyzed to determine the number of donor-origin
T cells per spleen that were represented in the proliferating vs the nonpro-
liferating group based on CFSE intensity. This was calculated for individ-
ual spleens by multiplying the percentage of cells determined in Fig. 8 by
the total number of leukocytes per spleen. The values represent the mean
cell numbers SD from three mice per group.
FIGURE 7. In vivo administration of IL-7 does not induce T cell acti-
vation markers or alter memory markers. C57BL/6 mice were injected i.p.
twice a day for 2 days with HBSS plus 0.1% NMS or IL-7 (10
g/injec-
tion). Peripheral lymph node cells were surface-labeled with fluorochrome-
conjugated mAb to CD4 and CD8 and mAb to the T cell activation markers
CD25, CD69, CD71, and CD137 and the memory markers CD44 and
CD62L. The histograms were generated by gating on either the CD4
or
CD8
T cell subset and displaying their expression of a particular activa
-
tion marker. The histogram of the control group is indicated by a dashed
line and that of the IL-7 group by a dark solid line. As a positive control
(light solid line) normal lymph node cells were activated in culture with
PMA and ionomycin or Con A, or anti-CD3 and anti-CD28, and labeled as
indicated above to demonstrate up-regulation of activation markers or al-
teration in memory markers.
FIGURE 8. In vivo administration of IL-7 increases the proportion of
donor T cells (CD45.1, IL-7R
) undergoing proliferation in IL-7R
/
re
-
cipient mice. Peripheral lymph node cells from C57BL/6-CD45.1 congenic
mice were labeled ex vivo with CFSE. C57BL/6-CD45.2
IL-7R
/
mice
were injected i.v. with 85 10
6
of the CFSE-labeled donor cells. These
donor cells contained 40.7% CD4
T cells and 37.8% CD8
T cells. After
allowing the injected cells 24 h to home to lymphoid tissues, mice were
injected i.p. twice a day for 7 days with HBSS plus 0.1% NMS (control)
or 10
g/injection of IL-7. Splenocytes were labeled with mAb specific to
CD45.1 and either CD4 or CD8 to distinguish donor-origin T cell subsets.
In addition, the cells were analyzed to determine the intensity of CFSE as
an indicator of proliferation. The histograms display the intensity of CFSE
of donor-origin CD4
T cells or CD8
T cells. This is a representative
profile of three mice per group. The histograms of the control group or the
IL-7-treated group are indicated by a dashed or solid line, respectively.
3025The Journal of Immunology
by IL-7 pretreatment in vivo. Indeed, the results presented here are
the first to demonstrate that IL-7 treatment in vivo induces Cdk2
kinase activity and Rb phosphorylation and increases cyclin E lev-
els; components involved in the G
1
/S transition. This complements
previous work by Itoh et al. (25) demonstrating that IL-7 increased
Cdk4 kinase activity in B cell precursors. Thus, IL-7 up-regulates
the cell cycle machinery, thereby allowing T cells from IL-7-
treated mice to undergo proliferation in an enhanced, accelerated
fashion after exposure to a subsequent stimulation.
The increased basal proliferation induced by IL-7 appears to be
more of a “homeostatic” proliferation rather than an “activation”
proliferation in that these T cells do not produce cytokine and they
do not express the markers CD25, CD69, CD71 (except for the
CD8
T cells), and CD137, which indicate activation. This is in
contrast to previous in vitro reports demonstrating that IL-7 in-
duces several activation markers, suggesting a state of activation
(3, 8, 11, 26). In addition, our results demonstrate that 2 days of
IL-7 treatment does not alter the expression of the activation/mem-
ory markers CD44 or CD62L (27–29) on either T cell subset,
suggesting that IL-7 does not activate naive cells or induce them to
become memory cells. Basal proliferation levels increase by day 2
in both T cell subsets, but occur disproportionately in the CD8
T
cells by a 3-fold greater amount, suggesting that IL-7 may have a
differential effect on CD8
T cells compared with CD4
T cells.
The up-regulation of CD71 only on the CD8
T cells and dispro
-
portionate increase in the number of CD8
T cells after 7 days of
IL-7 administration reported previously (15) support this hypoth-
esis. Although the disproportionate increase in the basal prolifer-
ation level of CD8
T cells could be attributable to the observation
that a higher percentage of CD8
T cells express the IL-7R com
-
pared with the CD4
T cell subset (30), this would not explain the
differential expression of CD71 on CD8
T cells compared with
the CD4
T cells with IL-7 treatment in vivo. Although these
results may not necessarily represent a role for IL-7 in normal
immune system homeostasis, it appears that exogenous adminis-
tration of IL-7 increases basal proliferation of T cells and enhances
functional capacity via a homeostatic mechanism. This may be
beneficial in the clinical setting in that IL-7 primes the T cells for
enhanced functional activity, but this potential is not realized un-
less the T cells are specifically activated. Thus, IL-7 therapy in
patients would avoid massive polyclonal activation of the T cell
population while concurrently enhancing responses to specific
stimuli such as peptides derived from tumor-associated Ags.
IL-7 administration has been shown previously to increase T
cell numbers in vivo (13–15). There are several possible ways that
this could be accomplished, including redistribution of T cells,
increased exportation from the thymus, expansion of peripheral T
cells by induction of proliferation often via activation, generation
of new T cells directly from precursors/progenitors via an extra-
thymic mechanism, and/or inhibition of apoptosis. It is unlikely
that the increase in T cell number is due to redistribution in that T
cell numbers are increased in all secondary lymphoid tissues ex-
amined (13–15). In addition, the increase in T cell numbers is not
attributable to increased exportation from the thymus, as demon-
strated by a previous report from our laboratory showing that T
cell numbers are increased to similar levels in both thymectomized
and normal euthymic mice given IL-7 in vivo (15). In this report,
we have examined the possibility that IL-7 increases T cell num-
bers via induction of proliferation. The data presented here dem-
onstrate that IL-7 increases basal proliferation in both T cell sub-
sets and disproportionately in CD8
T cells. Thus, we speculated
that the increase in T cell number and the alteration in the CD4:
CD8 T cell ratio that occurs with IL-7 administration in vivo could
be attributable to increased homeostatic proliferation of peripheral
T cells. Our results in Figs. 8 and 9 indicate that IL-7 does indeed
expand T cell numbers, at least in part, by inducing additional T
cells to undergo proliferation and not merely by increasing the
number of divisions of already proliferating cells. Because the host
mice in our studies had disrupted IL-7R, only the injected donor
cells were physically capable of responding to IL-7. Thus, our data
demonstrate that IL-7 acts directly on T cells to increase their
numbers and not through an indirect mechanism. Moreover, this
rules out the possibility that the increase in T cell numbers is
attributable to the generation of new cells from progenitors via a
thymic-independent mechanism. This is based on the fact that
host-origin progenitors are unable to respond to IL-7, as they have
no functional IL-7R. In addition, leukocytes from the peripheral
lymph node used as donor-origin cells contain virtually no pro-
genitor/precursor cells. IL-7 also has been shown to inhibit apo-
ptosis (1). The data in this report rule out the possibility that the
increase in T cell numbers induced by IL-7 is due solely to the
accumulation of T cells by blocking cell turnover via an antiapop-
tosis mechanism. However, we speculate that the antiapoptotic ef-
fect of IL-7 may still be involved. Specifically, we hypothesize that
after the induction of proliferation by IL-7, the antiapoptotic ef-
fects of IL-7 may serve to maintain the survival of the proliferating
cells. This hypothesis fits with our previous results showing that T
cell numbers begin to return to normal levels by 1–2 wk following
cessation of IL-7 treatment in vivo (31). Further investigation is
necessary to address this issue.
These results demonstrate that in vivo IL-7 pretreatment in-
creases T cell numbers (particularly CD8
T cells) by increasing
basal proliferation via a nonactivating rather than an activation
type of mechanism. This increase in basal proliferation correlates
with the ability of T cells to respond in an enhanced fashion to a
subsequent proliferative stimulus as the cell cycle machinery is in
place from induction by IL-7 and may serve to potentiate Ag-
specific responses as indicated previously by in vitro studies (6, 7).
Moreover, the data clearly show that IL-7 has differential effects on
T cell subsets in that CD71 is up-regulated only on the CD8
T
cells and the increase in basal proliferation is more profound in this
subset as well. These results may be important for the clinical
development of IL-7 as a vaccine adjuvant or to ameliorate im-
munosuppression in that IL-7 expands T cell numbers and en-
hances their functional capacity via a nonactivating mechanism.
Thus, although IL-7 induces polyclonal priming of T cells, this
potential can be restricted to selected cells that can specifically
respond to a particular Ag of interest.
Acknowledgments
We thank Drs. Robert Wiltrout, John Ortaldo, and Scott Durum for critical
review of this manuscript; Tim Back, Kathy McCormick, Erin Parsoneault,
and John Wine for their excellent technical expertise; and Susan Charbon-
neau and Joyce Vincent for typing and editing this manuscript.
References
1. Appasamy, P. M. 1999. Biological and clinical implications of interleukin-7 and
lymphopoiesis. Cytokines Cell Mol. Ther. 5:25.
2. Morrissey, P. J., R. G. Goodwin, R. P. Nordan, D. Anderson, K. H. Grabstein,
D. Cosman, J. Sims, S. Lupton, B. Acres, and S. G. Reed. 1989. Recombinant
interleukin 7, pre-B cell growth factor, has costimulatory activity on purified
mature T cells. J. Exp. Med. 169:707.
3. Armitage, R. J., A. E. Namen, H. M. Sassenfeld, and K. H. Grabstein. 1990.
Regulation of human T cell proliferation by IL-7. J. Immunol. 144:938.
4. Londei, M., A. Verhoef, C. Hawrylowicz, J. Groves, P. De Berardinis, and
M. Feldmann. 1990. Interleukin 7 is a growth factor for mature human T cells.
Eur. J. Immunol. 20:425.
5. Grabstein, K. H., A. E. Namen, K. Shanebeck, R. F. Voice, S. G. Reed, and
M. B. Widmer. 1990. Regulation of T cell proliferation by IL-7. J. Immunol.
144:3015.
6. Kos, F. J., and A. Mullbacher. 1992. Induction of primary anti-viral cytotoxic T
cells by in vitro stimulation with short synthetic peptide and interleukin-7. Eur.
J. Immunol. 22:3183.
3026 IL-7 INCREASES T CELL NUMBERS AND FUNCTION WITHOUT ACTIVATION
7. Kos, F. J., and A. Mullbacher. 1993. IL-2-independent activity of IL-7 in the
generation of secondary antigen-specific cytotoxic T cell responses in vitro. J. Im-
munol. 150:387.
8. Welch, P. A., A. E. Namen, R. G. Goodwin, R. Armitage, and M. D. Cooper.
1989. Human IL-7: a novel T cell growth factor. J. Immunol. 143:3562.
9. Costello, R., H. Brailly, F. Mallet, C. Mawas, and D. Olive. 1993. Interleukin-7
is a potent co-stimulus of the adhesion pathway involving CD2 and CD28 mol-
ecules. Immunology 80:451.
10. Soares, M. V., N. J. Borthwick, M. K. Maini, G. Janossy, M. Salmon, and
A. N. Akbar. 1998. IL-7-dependent extrathymic expansion of CD45RAT cells
enables preservation of a naive repertoire. J. Immunol. 161:5909.
11. Fukui, T., K. Katamura, N. Abe, T. Kiyomasu, J. Iio, H. Ueno, M. Mayumi, and
K. Furusho. 1997. IL-7 induces proliferation, variable cytokine-producing ability
and IL-2 responsiveness in naive CD4
T-cells from human cord blood. Immu
-
nol. Lett. 59:21.
12. Risdon, G., J. Gaddy, F. B. Stehman, and H. E. Broxmeyer. 1994. Proliferative
and cytotoxic responses of human cord blood T lymphocytes following alloge-
neic stimulation. Cell. Immunol. 154:14.
13. 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.
14. Faltynek, C. R., S. Wang, D. Miller, E. Young, L. Tiberio, K. Kross, M. Kelley,
and E. Kloszewski. 1992. Administration of human recombinant IL-7 to normal
and irradiated mice increases the numbers of lymphocytes and some immature
cells of the myeloid lineage. J. Immunol. 149:1276.
15. Komschlies, K. L., T. A. Gregorio, M. E. Gruys, T. C. Back, C. R. Faltynek, and
R. H. Wiltrout. 1994. Administration of recombinant human IL-7 to mice alters
the composition of B-lineage cells and T cell subsets, enhances T cell function,
and induces regression of established metastases. J. Immunol. 152:5776.
16. Goodwin, R. G., S. Lupton, A. Schmierer, K. J. Hjerrild, R. Jerzy, W. Clevenger,
S. Gillis, D. Cosman, and A. E. Namen. 1989. Human interleukin 7: molecular
cloning and growth factor activity on human and murine B-lineage cells. Proc.
Natl. Acad. Sci. USA 86:302.
17. Wells, A. D., H. Gudmundsdottir, and L. A. Turka. 1997. Following the fate of
individual T cells throughout activation and clonal expansion: signals from T cell
receptor and CD28 differentially regulate the induction and duration of a prolif-
erative response. J. Clin. Invest. 100:3173.
18. Graziano, M., Y. St Pierre, C. Beauchemin, M. Desrosiers, and
E. F. Potworowski. 1998. The fate of thymocytes labeled in vivo with CFSE. Exp.
Cell Res. 240:75.
19. Leo, O., M. Foo, D. H. Sachs, L. E. Samelson, and J. A. Bluestone. 1987. Iden-
tification of a monoclonal antibody specific for a murine T3 polypeptide. Proc.
Natl. Acad. Sci. USA 84:1374.
20. Damia, G., K. L. Komschlies, C. R. Faltynek, F. W. Ruscetti, and R. H. Wiltrout.
1992. Administration of recombinant human interleukin-7 alters the frequency
and number of myeloid progenitor cells in the bone marrow and spleen of mice.
Blood 79:1121.
21. Pierres, A., P. Naquet, A. Van Agthoven, F. Bekkhoucha, F. Denizot, Z. Mishal,
A. M. Schmitt-Verhulst, and M. Pierres. 1984. A rat anti-mouse T4 monoclonal
antibody (H129.19) inhibits the proliferation of Ia-reactive T cell clones and
delineates two phenotypically distinct (T4
, Lyt-2, 3
, and T4
, Lyt-2, 3
)
subsets among anti-Ia cytolytic T cell clones. J. Immunol. 132:2775.
22. Shen, F. W. 1981. Monoclonal antibodies to mouse lymphocyte differentiation
alloantigens. In Monoclonal Antibodies and T Cell Hybridomas: Perspectives
and Technical Advances. G. J. Hammerling, U. Hammerling, and J. F. Kearney,
eds. Elsevier/North Holland and Biomedical Press, Amsterdam, pp. 25–31.
23. Samaridis, J., G. Casorati, A. Traunecker, A. Iglesias, J. C. Gutierrez, U. Muller,
and R. Palacios. 1991. Development of lymphocytes in interleukin 7-transgenic
mice. Eur. J. Immunol. 21:453.
24. 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.
25. Itoh, N., M. Yasunaga, M. Hirashima, O. Yoshida, and S. Nishikawa. 1995. Role
of IL-7 and KL in activating molecules controlling the G
1
/S transition of B
precursor cells. Int. Immunol. 8:317.
26. Yssel, H., P. V. Schneider, and L. L. Lanier. 1993. Interleukin-7 specifically
induces the B7/BB1 antigen on human cord blood and peripheral blood T cells
and T cell clones. Int. Immunol. 5:753.
27. Budd, R. C., J. C. Cerottini, C. Horvath, C. Bron, T. Pedrazzini, R. C. Howe, and
H. R. MacDonald. 1987. Distinction of virgin and memory T lymphocytes: stable
acquisition of the Pgp-1 glycoprotein concomitant with antigenic stimulation.
J. Immunol. 138:3120.
28. Jung, T. M., W. M. Gallatin, I. L. Weissman, and M. O. Dailey. 1988. Down-
regulation of homing receptors after T cell activation. J. Immunol. 141:4110.
29. Lee, W. T., and E. S. Vitetta. 1991. The differential expression of homing and
adhesion molecules on virgin and memory T cells in the mouse. Cell Immunol.
132:215.
30. Sudo, T., S. Nishikawa, N. Ohno, N. Akiyama, M. Tamakoshi, and H. Yoshida.
1993. Expression and function of the interleukin 7 receptor in murine lympho-
cytes. Proc. Natl. Acad. Sci. USA 90:9125.
31. Komschlies, K. L., T. A. Gregorio, T. T. Back, M. E. Gruys, G. Damia,
C. R. Faltynek, and R. H. Wiltrout. 1994. Effects of rhIL-7 on leukocyte subsets
in mice: implications for antitumor activity. In Tumor Immunology and Cancer
Therapy. R. H. Goldfarb and T. L. Whiteside, eds. Marcel Dekker, New York, pp.
95–104.
3027The Journal of Immunology
... T-cell proliferation and T-cell numbers increased following IL-7 treatment, which was enhanced with the addition of sCD127 with a greater increase in CD8 + Tcell number compared with that of CD4 + T-cells. These differences between CD4 + and CD8 + T-cells are consistent with reports by Geiselhart et al. and Sportès et al., 22,23 but the mechanisms responsible for these differences are unknown. There might be distinct underlying factors regulating the homeostatic proliferation of CD8 + and CD4 + T-cells in response to IL-7. ...
... 29 In the present study, T-cell subsets were distinguished by expression of CD62L and CD44. 22 In healthy mice, IL-7 treatment resulted in a decrease of CD62L high CD44 low CD4 + Tn cells and an increase of CD62L low CD44 high CD4 + effector T-cells in the spleen ( Figure 3A). And while IL-7 also induced a decrease of CD8 + Tn cells (CD62L high CD44 low ), this was associated with an increase of Tcm-like phenotype (CD62L high CD44 high ) ( Figure 3B). ...
... This appears to contrast with a previous study where IL-7 treatment did not induce Tn-to-Tcm differentiation in healthy mice. 22 As the effects of IL-7 on T-cells are reversible after treatment cessation ( Figure 1C) 30 it is certainly possible that differentiated T-cells in our model would revert to pretreatment Tn phenotype. ...
Article
Full-text available
Introduction Soluble forms of cytokine receptors can be involved in the endogenous regulation of cytokine activity. Soluble interleukin 7 receptor α (sCD127) naturally binds IL-7, therefore there is interest in its potential application as an immunotherapeutic agent to regulate IL-7. With the hypothesis that sCD127 enhances IL-7 activity, thus promoting T-cell proliferation in vivo, we sought to assess the effect of sCD127, IL-7 or IL-7 + sCD127 treatment on CD4⁺ and CD8⁺ T-cells in the blood and spleen of mice. Methods Peripheral blood mononuclear cells and splenocytes were prepared, and analyzed for T-cell number, phenotype and proliferation (Ki67⁺) by flow cytometry. Results IL-7 treatment induced T-cell proliferation, increased T-cell number, and triggered T-cell differentiation each of which was enhanced with the addition of sCD127. IL-7 + sCD127 treatment significantly increased spleen weight over that seen with IL-7 treatment alone. More pronounced proliferation and a greater increase in cell number was observed in CD8⁺ T-cells relative to the effect on CD4⁺ T-cells. Conclusions These findings suggest that the addition of sCD127 enhances IL-7-mediated T-cell proliferation and suggests a potential therapeutic use for sCD127.
... Studies have indicated that IL-7 and CCL19 play important roles in the maintenance and formation of the T-zone in lymphoid organs (21,22). IL-7, a non-hematopoietic cell-derived cytokine critical for the development of the immune system, is a major regulator of proliferation and homeostasis of CD8 and CD4 T cells (23)(24)(25). CCL19, which is constitutively expressed by stromal cells in the lymphoid T-zone (26,27), is a chemotactic agent for dendritic cells and T cells migrating to secondary lymphoid tissue and plays an important role in the initiation of the adaptive immune response (28). ...
... Our BCMA-7×19 CAR vector contained a binding domain that recognizes BCMA, a CD8 transmembrane region, an intracellular 4-1BB costimulatory molecule, and a CD3ζ T cell signaling domain. IL-7 regulates the proliferation of T cells and maintains the stability of the intracellular environment (25), and CCL19 is a chemotactic agent for recruiting CCR7 + T cells and dendritic cells (22,28,49,50). We showed the superiority of our BCMA-7×19 CAR-T cells in terms of proliferation and chemotaxis by calculating absolute cell counts and performing transwell migration assay, which provided a good start-up for solving the problems such as CAR-T cell proliferation, infiltration and accumulation in the tumor microenvironment. ...
Article
Full-text available
Chimeric antigen receptor (CAR) technology has revolutionized cancer treatment, particularly in malignant hematological tumors. Currently, the BCMA-targeted second-generation CAR-T cells have showed impressive efficacy in the treatment of refractory/relapsed multiple myeloma (R/R MM), but up to 50% relapse remains to be addressed urgently. Here we constructed the BCMA-targeted fourth-generation CAR-T cells expressing IL-7 and CCL19 (i.e., BCMA-7 × 19 CAR-T cells), and demonstrated that BCMA-7 × 19 CAR-T cells exhibited superior expansion, differentiation, migration and cytotoxicity. Furthermore, we have been carrying out the first-in-human clinical trial for therapy of R/R MM by use of BCMA-7 × 19 CAR-T cells (ClinicalTrials.gov Identifier: NCT03778346), which preliminarily showed promising safety and efficacy in first two enrolled patients. The two patients achieved a CR and VGPR with Grade 1 cytokine release syndrome only 1 month after one dose of CAR-T cell infusion, and the responses lasted more than 12-month. Taken together, BCMA-7 × 19 CAR-T cells were safe and effective against refractory/relapsed multiple myeloma and thus warranted further clinical study.
... Moreover, IL7 has been shown to play an important role in the development and preservation of memory immune cell function. Previous work has demonstrated that IL7 can induce naive T-cells to differentiate into a memory phenotype [20,21], and promote the proliferation and activity of effector T-cells against cancer [22][23][24]. In addition, suppressive regulatory T-cells (Tregs) have been shown to express relatively low levels of the IL7 receptor (IL7R) [25]. ...
Article
Full-text available
Clinically approved head and neck squamous cell carcinoma (HNSCC) immunotherapies manipulate the immune checkpoint blockade (ICB) axis but have had limited success outside of recurrent/metastatic disease. Interleukin-7 (IL7) has been shown to be essential for effector T-cell survival, activation, and proliferation. Here, we show that IL7 in combination with radiotherapy (RT) is effective in activating CD8 + T-cells for reducing tumor growth. Our studies were conducted using both human papillomavirus related and unrelated orthotopic HNSCC murine models. Immune populations from the tumor, draining lymph nodes, and blood were compared between treatment groups and controls using flow cytometry, proteomics, immunofluorescence staining, and RNA sequencing. Treatment with RT and IL7 (RT + IL7) resulted in significant tumor growth reduction, high CD8 T-cell tumor infiltration, and increased proliferation of T-cell progenitors in the bone marrow. IL7 also expanded a memory-like subpopulation of CD8 T-cells. These results indicate that IL7 in combination with RT can serve as an effective immunotherapy strategy outside of the conventional ICB axis to drive the antitumor activity of CD8 T-cells.
... Mature T cells can be categorised as CD4 + T cells or CD8 + T cells according to their CD molecular phenotype. Studies have shown that the ratio of CD4+/CD8 + is related to IL-2 [26,27], IL-7 [28] and IL-16 [29]. CD4 + T cells and CD8 + T cells have different functions, which play synergistic or restrictive roles, and the ratio of CD4 + /CD8 + T cells reflects changes in immune regulation. ...
Article
Full-text available
Background Patients with polycystic ovary syndrome (PCOS) exhibit a chronic inflammatory state, which is often accompanied by immune, endocrine, and metabolic disorders. Clarification of the pathogenesis of PCOS and exploration of specific biomarkers from the perspective of immunology by evaluating the local infiltration of immune cells in the follicular microenvironment may provide critical insights into disease pathogenesis. Methods In this study, we evaluated immune cell subsets and gene expression in patients with PCOS using data from the Gene Expression Omnibus database and single-sample gene set enrichment analysis. Results In total, 325 differentially expressed genes were identified, among which TMEM54 and PLCG2 (area under the curve = 0.922) were identified as PCOS biomarkers. Immune cell infiltration analysis showed that central memory CD4⁺ T cells, central memory CD8⁺ T cells, effector memory CD4⁺ T cells, γδ T cells, and type 17 T helper cells may affect the occurrence of PCOS. In addition, PLCG2 was highly correlated with γδ T cells and central memory CD4⁺ T cells. Conclusions Overall, TMEM54 and PLCG2 were identified as potential PCOS biomarkers by bioinformatics analysis. These findings established a basis for further exploration of the immunological mechanisms of PCOS and the identification of therapeutic targets.
... In this study, only the FSI model showed elevated RANTES levels, supporting the sepsis-like characteristics of the FSI model rather than the CLP model. IL-7 is a potent anti-apoptotic cytokine that is essential for lymphocyte survival and expansion (24,25). Only the CLP model showed significantly higher IL-7 levels in our study, but there was no correlation between plasma IL-7 levels and the severity of sepsis (26). ...
Article
Full-text available
Background Various animal models of sepsis have been developed to optimize sepsis treatment. However, therapeutic agents that were successful in animal models were rarely effective in human clinical trials. The cecal ligation and puncture (CLP) model is currently the gold standard for sepsis studies. However, its limitations include the high variability among researchers and the difficulty in comparing animals with different cecum shapes and sizes. In this study, we established a protocol for the creation of a simple and accessible sepsis rodent model using fecal suspensions that minimized differences in technical effects among researchers and individual differences in animals. Methods A mouse model of sepsis using fecal suspension intraperitoneal injection (FSI) was created using fresh stool excreted within 24 h. The collected fresh stool was dissolved in saline solution and filtered. The obtained fecal suspension was injected intraperitoneally into the mice. Moreover, fecal suspensions with different concentrations were prepared, and the survival rates were compared among the fecal suspensions for each concentration. To assess the validity of the FSI as a sepsis model, CLP and FSI with similar mortality rates were compared pathologically, physiologically, immunologically, and bacteriologically. Histopathological comparison was evaluated by hematoxylin-eosin and Gram staining of the parenchymal organs. Physiological evaluation was performed by comparing the respiratory rate, body temperature, and blood gas analysis results. Immunological assessment was performed using multiplex analysis. Bacteriological comparisons were performed by culturing ascites fluid. Results The FSI model increased mortality in proportion to the fecal suspension concentration. The mortality rate was reduced with antibiotic administration. In various comparative experiments conducted using the FSI and CLP models, both models showed findings consistent with sepsis. Furthermore, the FSI model showed less variability among the individuals in each test. Conclusion This is the first detailed and accurate report of a protocol for creating a sepsis model using fecal suspension. The FSI model is a minimally invasive and accessible sepsis rodent model. Its clinical validity as a sepsis model was proven via histological, physiological, microbiological, and immunological evaluation methods. The FSI model minimizes individual differences between mice and helps to conduct accurate studies after the onset of sepsis.
... IL7 is a 25-kDa glycoprotein that is required for human T-cell development and maintaining the homeostasis of mature T cells (11). IL7 is the main homeostatic driver of T-cell numbers (12). IL7 signals through the IL7 receptor (IL7R) and the downstream JAK-STAT pathway (11). ...
Article
Purpose: Patients with glioblastoma (GBM) are treated with radiation therapy (RT) and temozolomide (TMZ). These treatments may cause prolonged systemic lymphopenia, which itself is associated with poor outcomes. NT-I7 is a long-acting IL-7 that expands CD4 and CD8 T cell numbers in humans and mice. We tested whether NT-I7 prevents systemic lymphopenia and improves survival in mouse models of GBM. Experimental design: C57BL/6 mice bearing intracranial tumors (GL261 or CT2A) were treated with RT (1.8 Gy/day x 5 days), TMZ (33 mg/kg/day x 5 days), and/or NT-I7 (10 mg/kg on the final day of RT). We followed the mice for survival while serially analyzing levels of circulating T lymphocytes. We assessed regulatory T cells (Treg) and cytotoxic T lymphocytes in the tumor microenvironment, cervical lymph nodes, spleen, and thymus; and hematopoietic stem and progenitor cells (HSPCs) in the bone marrow. Results: GBM tumor-bearing mice treated with RT+NT-I7 increased T lymphocytes in the lymph nodes, thymus, and spleen, enhanced IFNγ production, and decreased Treg cells in the tumor which was associated with a significant increase in survival. NT-I7 also enhanced central memory and effector memory CD8 T cells in lymphoid organs and tumor. Depleting CD8 T cells abrogated the effects of NT-I7. Furthermore, NT-I7 treatment decreased progenitor cells in the bone marrow. Conclusion: In orthotopic glioma-bearing mice, NT-I7 mitigates radiation-related lymphopenia, increases cytotoxic CD8 T lymphocytes systemically and in the tumor, and improves survival. A phase I/II trial to evaluate NT-I7 in patients with high-grade gliomas is ongoing (NCT03687957).
... However, IL7 induce different effects in CD4+ and CD8+ T-cells depending on the context. For instance, IL7 exposure of resting CD4+ T-cells do not induce cell proliferation however, IL7 induces a proliferative advantage on CD4+ Tcells engaged into proliferation via other stimuli (Ponchel et al., 2005;Geiselhart et al., 2001). Proliferating CD4+ T-cells may also gain survival benefit from engaging IL7, however only at a time when such survival depends on external signals to persist (i.e. ...
Article
Interfering with signalling pathways by targeting cell surface proteins has become an important strategy in the development of novel therapeutic agents. Notably, interfering with cytokine signalling revolutionised the treatment of chronic diseases. Cytokines can induce a range of effects that are not always accounted for in assays detecting cytokine binding to cell surface receptors and/or proximal signalling interference. Hence, robust assays are needed to characterise the activity of potential drug candidates targeting such effects. We chose interleukin-7 (IL7) as a cytokine model due to its long-term effect on T-cells. In this report we describe the development and refinement of an in vitro assay for measuring the long-term effect of IL7, more specifically on CD4+ T-cells, while the assay could be adapted to look at CD8+ T-cells. PBMCs and/or purified CD4+ T-cells stained with VPD450 (cell cycle dye) were expanded for 5 days using the mitogen Phytohemagglutinin and/or CD3/CD28 agonists. This resulted in cell proliferation (VPD450 dilution) and activation-induced cell death (7-AAD uptake) which was rescued by the addition of IL7, resulting in cell survival over a further 5 days. JAK-inhibitor (Tofactinib) or a blocking anti-IL7Rα antibody (clone R34.34) abolished cell survival suggesting antagonism, while another antibody (clone A019D5) displayed an agonist effect. These results were confirmed at the proximal signalling level using an IL7/STAT5-luciferase reporter assay. This novel assay for a biological long term effect may be useful for the characterisation of potential therapeutic drugs targeting the IL7/IL7R in CD4+ T-cells.
Article
Full-text available
Interleukin-7 (IL-7) is a versatile cytokine that plays a crucial role in regulating the immune system’s homeostasis. It is involved in the development, proliferation, and differentiation of B and T cells, as well as being essential for the differentiation and survival of naïve T cells and the production and maintenance of memory T cells. Given its potent biological functions, IL-7 is considered to have the potential to be widely used in the field of anti-tumour immunotherapy. Notably, IL-7 can improve the tumour microenvironment by promoting the development of Th17 cells, which can in turn promote the recruitment of effector T cells and NK cells. In addition, IL-7 can also down-regulate the expression of tumour growth factor-β and inhibit immunosuppression to promote anti-tumour efficacy, suggesting potential clinical applications for anti-tumour immunotherapy. This review aims to discuss the origin of IL-7 and its receptor IL-7R, its anti-tumour mechanism, and the recent advances in the application of IL-7 in tumour therapy.
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
Interleukin-7 (IL-7) is a cytokine belonging to the chemokine family. It plays a key role in the differentiation, development, and maturation of T lymphocytes and B lymphocytes, which is pivotal to adaptive immunity. In addition to its role in lymphocyte development, recent studies have indicated the antitumor functions of IL-7 in the tumor microenvironment. In this review, we discuss the role of IL-7 in tumors and summarize its antitumor potential and clinical application in lymphoma, leukemia, breast cancer, colon cancer, and so on. Furthermore, the combinational strategies of IL-7 and other antitumor drugs have been also discussed.
Article
Full-text available
The administration of greater than or equal to 5 micrograms interleukin- 7 (IL-7) twice a day to mice for 4 to 7 days increased by twofold to fivefold the total number of splenic and peripheral blood leukocytes, but did not appreciably increase bone marrow (BM) cellularity. This regimen of IL-7 administration also resulted in a greater than 90% reduction in the frequency and total number of single lineage colony- forming unit-culture (CFU-c) and multilineage CFU-granulocyte, erythroid, monocyte, megakaryocyte colonies that could be cultured from the BM, but a fivefold to 15-fold increase in the number of these progenitors that could be cultured from the spleen. All of these effects were reversible with progenitor and white blood cell numbers returning to near normal by day 6. Morphologic analysis of cells obtained from the BM of IL-7-treated mice showed an increase in lymphoid cells. Surface phenotype analysis showed that most of this IL- 7-induced increase in lymphocytes was attributable to an increase in immature B cells (B220+, sIg-), while cells expressing the myelomonocytic markers 8C5 and MAC-1 decreased by twofold to threefold. Further studies showed that the administration of IL-7 to mice that had been rendered leukopenic by the injection of cyclophosphamide (Cy) or 5- fluorouracil (5FU) exhibited a more rapid recovery and/or overshoot in their peripheral blood lymphocytes when compared with mice treated with Cy or 5FU alone. These results show that IL-7 can differentially regulate myelopoiesis in the BM and spleen, while stimulating lymphopoiesis.
Article
Interleukin 7 (IL-7) is a stromal cell derived cytokine that stands out as being the only cytokine identified to date on which development of B and T lymphocytes is absolutely dependent. IL-7 functions primarily as a growth and anti-apoptosis factor for B- and T cell (alpha beta and gamma delta TCR+ cells) precursors, and is essential for differentiation of gamma delta TCR+ cells. IL-7 can function as a cofactor during myelopoiesis, and is capable of activating monocytes/macrophages and natural killer (NK) cells. Its receptor (IL-7R) is a heterodimer of an alpha chain that specifically binds IL-7 and the common gamma chain gamma(c) that is also a component of the receptors for IL-2, IL-4, IL-9 and IL-15. The functions of IL-7 in normal lymphocyte development and activation have led to the demonstration of the ability of IL-7 to stimulate lymphopoiesis in lymphopenic mice, suggesting a possible clinical application of IL-7 in accelerating lymphoid reconstitution in lymphopenic patients. There have also been a number of preclinical studies pointing to the possible utility of IL-7 in antitumor clinical applications, and clinical trials involving IL-7 gene therapy of metastatic disease are underway. IL-7 has also been shown to promote engraftment of stem cells in mice receiving bone marrow transplants, pointing to a possible use of IL-7 in patients receiving bone marrow or peripheral blood stem cell transplants. Areas of IL-7 biology that are essentially unexplored include the mechanisms of regulation of the expression of IL-7 and IL-7R alpha, as well as the mechanisms by which IL-7 is a growth and differentiation factor for gamma delta T cells but a growth factor only for alpha beta T cells.
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
While chemically defined conditions for culturing normal tissue have been attained for only a few cell types, the sustained proliferation of a precursor cells expressing IL-7 receptor and c-Kit can be supported under chemically defined conditions containing recombinant IL-7 and the ligand for c-Kit (KL). To understand the biochemical basis of the cell cycle progression of a precursor cells proliferating under these conditions, we investigated the correlation between growth factor stimulation and CDK4 activity. Consistent with our findings that IL-7 regulates the G(1)/S transition, while KL has only a little role in this process, the kinase activity of CDK4 was related closely with IL-7 stimulation but not KL stimulation. We investigated the mechanism underlying CDK4 activation in the IL-7-stimulated B precursor cells. Our results showed that (i) CDK4 and cyclin D3 are the G(1)/S regulators in B precursor cells; (ii) their expression levels are unchanged between the cells in G(1) arrest and cycling cells; and (iii) they are present in an associated form even when the cell cycle stage is arrested at G(1). Thus, the regulation of the expression of CDK4 and cyclin D3 or regulation of their assembly are not the mechanisms for activating CDK4 in the B precursor cells, On the other hand, a number of molecules co-immunoprecipitated with CDK4 were enhanced in the lysate of IL-7-stimulated B precursor cells. Thus, we present a possibility that CDK4 activation might be regulated by molecules associated with the CDK4-cyclin D3 complex in an IL-7-dependent manner.
Article
The present study investigated whether a short synthetic peptide NPP, with a modified sequence (147-158 R156-) derived from influenza A virus nucleoprotein with high affinity for Kd major histocompatibility complex class I molecules, could induce primary influenza virus-specific cytotoxic T (Tc) cells in vitro. Naive BALB/c (H-2d) splenocytes did not respond to the stimulation with only NPP with the generation of effector Tc cells specific for influenza A virus-infected target cells in vitro. However, they were able to do so if cultured with NPP in the presence of IL-7. IL-7 activity in this system differed significantly from IL-2 activity in the specificity of the effect. The use of exogenous IL-2, instead of IL-7, with NPP resulted in the induction of lytic cells that lysed both influenza virus-infected and uninfected syngeneic target cells. These results suggest that IL-7 is a potent regulatory cytokine in the antigen-specific activation of resting naive Tc cell precursors and may provide the necessary conditions for the induction of human primary Tc cells in vitro.
Article
In vitro experiments performed by several investigators have demonstrated that IL-7 is a growth factor for immature B lymphocytes, thymocytes, and mature T lymphocytes. To evaluate the potential therapeutic use for human rIL-7 (rhuIL-7) as a hematopoietin, we have studied the in vivo hematopoietic effects of rhuIL-7 in mice. In these experiments, sublethally irradiated and normal mice were treated with or without rhuIL-7 for up to 26 days. Administration of rhuIL-7 significantly increased the white blood cell count in the peripheral blood and spleen in both normal and irradiated mice. Treatment with rhuIL-7 also accelerated lymphocytic recovery in irradiated mice. Precursor and mature B lymphocytes showed the greatest expansion in response to rhuIL-7 administration, with smaller increases in T lymphocytes being observed. In mice recovering from high dose irradiation, rhuIL-7 treatment resulted in preferential expansion of CD8+ T lymphocytes and more rapid normalization of the CD4/CD8 ratios. Differential analysis of peripheral blood smears demonstrated that rhuIL-7 also increased the numbers of immature granulocytes in both normal and irradiated mice. Moreover, administration of rhuIL-7 to normal, irradiated, cyclophosphamide-pretreated, or 5-fluorouracil-pretreated mice increased the number of acetylcholinesterase-positive megakaryocytes in the spleen, but not the bone marrow. Therefore, although the major in vivo effects of rhuIL-7 were on cells of the lymphocytic lineage, rhuIL-7 also increased the numbers of some immature cells of the myeloid lineage.
Article
The administration of greater than or equal to 5 micrograms interleukin-7 (IL-7) twice a day to mice for 4 to 7 days increased by twofold to fivefold the total number of splenic and peripheral blood leukocytes, but did not appreciably increase bone marrow (BM) cellularity. This regimen of IL-7 administration also resulted in a greater than 90% reduction in the frequency and total number of single lineage colony-forming unit-culture (CFU-c) and multilineage CFU-granulocyte, erythroid, monocyte, megakaryocyte colonies that could be cultured from the BM, but a fivefold to 15-fold increase in the number of these progenitors that could be cultured from the spleen. All of these effects were reversible with progenitor and white blood cell numbers returning to near normal by day 6. Morphologic analysis of cells obtained from the BM of IL-7-treated mice showed an increase in lymphoid cells. Surface phenotype analysis showed that most of this IL-7-induced increase in lymphocytes was attributable to an increase in immature B cells (B220+, sIg-), while cells expressing the myelomonocytic markers 8C5 and MAC-1 decreased by twofold to threefold. Further studies showed that the administration of IL-7 to mice that had been rendered leukopenic by the injection of cyclophosphamide (Cy) or 5-fluorouracil (5FU) exhibited a more rapid recovery and/or overshoot in their peripheral blood lymphocytes when compared with mice treated with Cy or 5FU alone. These results show that IL-7 can differentially regulate myelopoiesis in the BM and spleen, while stimulating lymphopoiesis.
Article
We have used an anti-CD45R mAb to identify virgin and memory CD4+ T cells. In this report, we demonstrate that in mice, as in humans, virgin and memory T cells express different levels of adhesion molecules. Splenic CD45Rlo memory T cells express higher levels of PgP-1, Lyt-1, and LFA-1 than do CD45Rhi virgin cells. In contrast, CD45Rlo memory cells express lower levels of antigens identified by the mAbs. Mel-14 (leukocyte homing receptor) and SM3G11. These observations are in agreement with work in humans and rats which suggests that differences in the levels of adhesion molecules and homing receptors might be related to the efficiency of activation and patterns of recirculation among virgin and memory T cells.
Article
Normal mice were injected with IL-7 (500 ng, twice daily) for various periods of time up to 6 days and the cellularity and phenotypic composition of the thymus, spleen, lymph node, and bone marrow was assessed. After 6 days of treatment, significant increases in the cellularity of the spleen, lymph node, and bone marrow were observed which returned to the normal range within 6 days after cessation of treatment. After 3 days of IL-7 treatment, increased numbers of B220+/surface(s) IgM- bone marrow cells were observed. After 6 days of treatment, these numbers were still further increased and a significant population of B220+/sIgM- cells were observed in the spleen. The numbers of c mu+/sIgM- cells were also increased in the IL-7-treated mice. Analysis of the expression of B220 and BP-1 on the sIgM- bone marrow cells revealed that the B220+/BP-1+ population was dramatically increased after IL-7 treatment and the size of the B220+/BP-1- population did not differ from control mice. The pre-B cell numbers declined rapidly after the cessation of IL-7 treatment. After 6 days of IL-7 treatment, a twofold increase in the number of B cells in the spleen and lymph node was observed. The B cell numbers declined to normal values within 6 days after the cessation of IL-7 administration. In the spleens of the IL-7-treated mice, there was a significant increase in the number of B cells with an immature phenotype (e.g., sIgMhi/sIgDlo, decreased levels of Ia and FcR expression). The numbers of CD8+ and CD4+ T cells were also increased in the lymph node and spleen of the IL-7-treated mice. These numbers declined to normal levels after the cessation of IL-7 treatment.
Article
We have studied the capacity of interleukin (IL) 7 to support the growth and expansion of human T cell clones of different phenotype and function. All the clones studied (CD4+CD8+, CD4-CD8- Tcell receptor alpha/beta or gamma/delta) responded to IL 7. The proliferative response of all the T cell clones induced by IL 7 was routinely less than to IL2, but comparable to the IL4 response. IL 7 also induced the proliferation of resting, freshly prepared peripheral blood mononuclear cells (PBMC) or Tcell-enriched (E+) cells. The pattern of proliferation observed in the presence of IL 7 was similar, but lower in magnitude, to that induced by IL 2. In both these cells populations the response to lymphokines alone was always less than the response to lymphokines plus insolubilized anti-CD3 monoclonal antibody. In contrast IL4 produced a different pattern of responsiveness, as significant proliferation was observed only on PBMC costimulated with anti-CD3. The possibility that IL 7 mediates its growth stimulation by the IL2 pathway was excluded by the incapacity of anti-IL2 or anti-Tac monoclonal antibody, in concentrations which blocked IL2-dependent proliferation, to inhibit IL 7-dependent growth. We conclude that IL 7 is a major growth factor for human mature T cells, and its activity is not limited to lymphocyte progenitors.