ArticlePDF Available

The Trophic Action of IL-7 on Pro-T Cells: Inhibition of Apoptosis of Pro-T1, -T2, and -T3 Cells Correlates with Bcl-2 and Bax Levels and Is Independent of Fas and p53 Pathways1

Authors:

Abstract and Figures

Signals from the IL-7R are essential for normal thymocyte development. We isolated thymocytes from early developmental stages and observed that suspensions of pro-T1, -T2, and -T3 cells rapidly died in culture. Addition of IL-7 promoted their survival, but did not induce cell division. Pro-T4 cells did not undergo rapid cell death, and their survival was therefore independent of IL-7. Death in the absence of IL-7 showed the hallmarks of apoptosis, including DNA fragmentation and annexin V binding; however, caspase inhibitors blocked DNA fragmentation, but did not block cell death. The trophic effect of IL-7 was partially inhibited by blocking protein synthesis. The p53 pathway was not involved in this death pathway, since pro-T cells from p53-/- mice also underwent cell death in the absence of IL-7. The Fas/Fas ligand pathway was not involved in cell death, since Fas-deficient pro-T cells died normally in the absence of IL-7, anti-Fas Abs did not protect cells from death in the absence of IL-7, and Fas expression was undetectable on cells at these stages. The IL-7 trophic affect correlated with increased intracellular levels of Bcl-2 and decreased levels of Bax, whereas no Bcl-X(L), Bcl-w, or Bad was detectable. Thus, maintaining a favorable Bcl-2/Bax ratio may account for the trophic action of IL-7.
Content may be subject to copyright.
The Trophic Action of IL-7 on Pro-T Cells: Inhibition of
Apoptosis of Pro-T1, -T2, and -T3 Cells Correlates with Bcl-2
and Bax Levels and Is Independent of Fas and p53 Pathways
Kyungjae Kim,* Chong-kil Lee,* Thomas J. Sayers,
Kathrin Muegge,
and Scott K. Durum*
Signals from the IL-7R are essential for normal thymocyte development. We isolated thymocytes from early developmental stages
and observed that suspensions of pro-T1, -T2, and -T3 cells rapidly died in culture. Addition of IL-7 promoted their survival, but
did not induce cell division. Pro-T4 cells did not undergo rapid cell death, and their survival was therefore independent of IL-7.
Death in the absence of IL-7 showed the hallmarks of apoptosis, including DNA fragmentation and annexin V binding; however,
caspase inhibitors blocked DNA fragmentation, but did not block cell death. The trophic effect of IL-7 was partially inhibited by
blocking protein synthesis. The p53 pathway was not involved in this death pathway, since pro-T cells from p53
2/2
mice also
underwent cell death in the absence of IL-7. The Fas/Fas ligand pathway was not involved in cell death, since Fas-deficient pro-T
cells died normally in the absence of IL-7, anti-Fas Abs did not protect cells from death in the absence of IL-7, and Fas expression
was undetectable on cells at these stages. The IL-7 trophic affect correlated with increased intracellular levels of Bcl-2 and
decreased levels of Bax, whereas no Bcl-X
L
, Bcl-w, or Bad was detectable. Thus, maintaining a favorable Bcl-2/Bax ratio may
account for the trophic action of IL-7. The Journal of Immunology, 1998, 160: 5735–5741.
N
ormal T cell development in the mouse and in man has
been shown to depend on signals from the IL-7R (re-
viewed in Ref. 1). This conclusion is based on the phe-
notypes of mice deficient for IL-7R
a
(2, 3), IL-7 itself (4, 5), mice
treated with Abs against IL-7 (6, 7), and deficiencies of the
g
c
component of the IL-7R in humans (8) and mice (9, 10).
Some
ab
T cell development occurs in about 40% of IL-7R
2/2
mice, suggesting that alternative pathways can also support T cell
development; however, the peripheral T cells that eventually ac-
cumulate in these leaky mice do not proliferate in response to
stimuli (11). The
gd
T cell lineage is completely undetectable in all
IL-7R
2/2
mice (3, 12) based on a failure to rearrange the TCR
g
locus (13, 14) or a failure to express these genes (15). The devel-
opment of B lymphocytes is also severely impaired in IL-7R
a
2/2
mice and
g
c
2/2
mice, but not in
g
c
-deficient humans reflecting the
IL-7 independence of human B cells (16). NK development is
normal in IL-7R
2/2
mice (12).
The earliest stages in murine T cell development (before ex-
pression of CD4, CD8, and CD3) have been distinguished based
on CD44 and CD25 expression (reviewed in Ref. 17): pro-T1
(CD44
1
CD25
2
), pro-T2 (CD44
1
CD25
1
), pro-T3 (CD44
2
CD25
1
),
and pro-T4 (CD44
2
CD25
2
). Expression of c-Kit, the receptor for
stem cell factor, corresponds with expression of CD44 at these
pro-T cell stages. The CD25
1
stages are deficient in IL-7R
2/2
mice and in mice treated with anti-IL-7. This suggests that one
requirement for IL-7R signals occurs before or during the pro-T2
stage. IL-7 has been reported to induce proliferation of early T
cells (18) and could therefore play a role in the expansion as well
as differentiation or survival of these cells. To clarify the nature of
the IL-7R signal requirement, pro-T cells at different stages were
isolated from the thymus and examined for the effects of IL-7 on
survival and cell cycle, and the cell death process that occurred in
the absence of IL-7 was characterized.
Materials and Methods
Mice
C57BL/6 and MRL-lpr/lpr mice were housed in a specific pathogen-free
environment. Mice were mated overnight and checked for plugs the fol-
lowing day, which was designated day 1 of gestation. On the indicated day
of gestation, mothers were killed by CO
2
asphyxiation, and embryos were
killed by chilling on ice. Thymi were removed from embryos using a dis-
secting microscope. Rag-2
2/2
(19) and p53
2/2
(20) mice were bred in our
facility from breeders purchased from The Jackson Laboratory (Bar
Harbor, ME).
Preparation and culture of fetal thymocytes
Fetal thymus lobes were obtained from embryos on day 14, 15, 16, 17, or
18 of gestation. Cell suspensions were prepared by gentle disruption with
a micropipette after treatment with 0.2% collagenase (Sigma, St. Louis,
MO) dissolved in PBS containing 20% heat-inactivated FCS (HyClone,
Logan, UT) for1hat37°C. The cells were cultured in RPMI 1640 sup-
plemented with 2 mM glutamine, 100 U/ml penicillin, 100
m
g/ml strepto-
mycin, 50
m
M 2-ME, and 10% heat-inactivated FCS. Fetal thymocytes
(4 3 10
5
/200
m
l/well) were cultured in 96-well U-bottom plates in the
presence or the absence of IL-7 (50 ng/ml; PeproTech, Rocky Hill, NJ) for
the indicated length of time. Hamster anti-Fas (Jo2; 10
m
g/ml; PharMin-
gen, San Diego, CA) was added to some cultures. Caspase inhibitors, z-
VAD-FMK and z-DEVD-FMK (20
m
M; Enzyme Systems Products, Dub-
lin, CA), were preloaded into thymocytes at 0°C for 30 min before placing
cells in culture together with inhibitors at 37°C.
Anti-Fas control treatment
Renca cells were stimulated with IFN-
g
(100 U/ml for 18 h) to increase
their susceptibility to fas-mediated killing. Renca cells were then labeled
with
51
Cr and incubated with d11S cells that express FasL
2
and hamster
anti-Fas (10
m
g/ml) for 16 h, and the release of
51
Cr was determined.
*Laboratory of Molecular Immunoregulation and
Science Applications International
Corporation, Division of Basic Sciences, National Cancer Institute, Frederick, MD
21702
Received for publication August 28, 1997. Accepted for publication February 9, 1998.
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
Address correspondence and reprint requests to Dr. Scott K. Durum, Laboratory of
Molecular Immunoregulation, National Cancer Institute, Building 560, Room 31-45,
Frederick Cancer Research Facility, Frederick, MD 21702-1201.
2
Abbreviations used in this paper: FasL, Fas ligand.
Copyright © 1998 by The American Association of Immunologists 0022-1767/98/$02.00
Flow cytometric analysis and cell sorting with Abs
For Ab staining, cells were harvested, washed in a staining solution of PBS
containing 5% FCS and 0.1% NaN
3
, and resuspended in 50
m
l of staining
solution containing 0.5
m
g of rat mAb 2.4G2 (anti-mouse Fc
g
RII, PharM-
ingen) and 10% normal mouse serum to reduce nonspecific binding of Abs
to Fc receptors. Cell suspensions were stained with mAbs (1/200 dilution)
for 20 min at 4°C. Abs were purchased from PharMingen, including R-
phycoerythrin-conjugated anti-CD44 (clone IM7), FITC-conjugated anti-
CD25 (clone 7D4), FITC-anti-CD4 (RM4-5), and R-phycoerythrin-conju-
gated anti-mouse CD8
a
(clone 53-6.7). Cells were washed and fixed in 1%
paraformaldehyde in PBS, and analyzed on a FACStar Plus (Becton Dick-
inson Immunocytometry System, Mountain View, CA), gating out dead
cells by forward low angle scatter and low right angle scatter. For sorting
cells, the same staining method was used without fixation, and cells were
sorted on a FACStar Plus or a modified FACSII (Becton Dickinson).
Pro-T1 (CD44
1
CD25
2
) cells were sorted from day 14 thymocytes. Pro-T2
(CD44
1
CD25
1
) cells were sorted from day 14 or 15 thymocytes. Pro-T3
(CD44
2
CD25
2
) cells were sorted from day 16 thymocytes or from
Rag2
2/2
adult thymocytes. Pro-T4 (CD4
2
CD8
2
CD44
2
CD25
2
) cells
were sorted from day 17 thymocytes. From p53
2/2
mice a mixture of
pro-T2 and pro-T3 cells (CD4
2
CD8
2
CD25
1
) was sorted from thymocytes
from adult mice. The purity of sorted populations was generally .98%.
Sorted cells were then cultured overnight with or without IL-7.
Intracellular staining of Bcl-2, Bcl-X
L
, Bax, and Bad
Single-cell suspensions were permeabilized with saponin buffer (PBS con-
taining 1% BSA (Sigma) and 0.04% saponin) for 20 min, then incubated
with monoclonal anti-mouse Bcl-2 (clone 3F11, PharMingen), anti-mouse
Bcl-X
L
(clone 4, Transduction Laboratory, Lexington, KY), anti-mouse
Bad (clone 2G11, PharMingen), or a polyclonal anti-mouse Bax (Santa
Cruz Biotechnology, Santa Cruz, CA) for 30 min on ice. Isotype-matched
control Abs were purified mouse IgG2b (clone 49.2 PharMingen), purified
polyclonal hamster IgG (PharMingen), or purified rabbit IgG. Cells were
washed twice with saponin buffer, incubated with FITC anti-hamster IgG
(clone G94-56, PharMingen), FITC anti-mouse IgG2b (clone R12-3
PharMingen), or FITC-anti-rabbit IgG (Santa Cruz Biotechnology) for 30
min at 4°C, and washed once with saponin buffer and once with PBS
containing 1% BSA.
Annexin V staining and viability
Annexin V binding was performed with a commercial kit (Clontech, Palo
Alto, CA). Cells were collected, then resuspended in annexin V binding
buffer containing annexin V-FITC (1
m
g/ml) and propidium iodide (2
m
g/
ml) for 10 min at room temperature in the dark, washed, and analyzed by
microfluorometry. To analyze for viability, cells were incubated with pro-
pidium iodide, then the percentage of cells excluding the dye was deter-
mined by flow microfluorometry.
Cell cycle analysis
Cells were placed in a detergent buffer (21) and an equal volume of staining
buffer (propidium iodide, 50
m
g/ml). Cells were mixed by inversion, in-
cubated at room temperature in the dark for 1 h, and analyzed by flow
microfluorometry.
Results
Thymocytes from mouse embryos on day 14 of gestation were
cultured with or without IL-7 for various times. The recovered
cells were examined for viability by exclusion of propidium io-
dide, which stains the nuclei of either necrotic or apoptotic cells.
Apoptotic cells were detected by binding annexin V to their
plasma membranes. As shown in Figure 1, these freshly explanted
thymocytes began to die by apoptosis within 2 h, and by 20 h, 90%
had undergone apoptotic death. IL-7 protected about half the cells
from apoptotic death at 20 h, and its survival effects could be seen
as early as 6 h. In .20 experiments, IL-7 protected 50 to 80% of
the cells from death at the 20 h point. Further culture in IL-7
beyond 20 h (not shown) showed that with each additional day,
about half of the cells died. We noted that culture density had an
effect on viability, with IL-7 being less effective as culture density
was lowered.
The thymocyte population on day 14 of gestation is comprised
of the first two stages of pro-T cell development, pro-T1 and pro-
T2, in approximately equal proportions. We sorted the cells into
pro-T1 and pro-T2 based on the CD25 and CD44 markers and
tested each population for the effect of IL-7 using cell cycle anal-
ysis. In this analysis, apoptotic cells were visualized based on
DNA fragmentation to subG1 quantities of DNA. As shown in
Figure 2, both pro-T1 and pro-T2 populations were protected from
apoptotic death by IL-7. We also visualized DNA fragmentation
by gel electrophoresis and observed a comparable IL-7 effect (not
shown). It is clear from this analysis that IL-7 did not promote
cycling of pro-T cells, as shown by the decline in the proportion of
cells in S and G
2
-M during culture in IL-7 compared with that at
the start of the culture (see also Fig. 6). The percentages of pro-T2
cells in G
1
, S, and G
2
-M phases when initially isolated were 56.5,
37.5, and 6.1%, respectively. After IL-7 culture, they were 81.5,
16.5, and 1.9%, respectively. Thus, cell division declined during
culture in IL-7, and its activity is best described as “trophic” rather
than “growth” for pro-T cells.
The later stages of pro-T cell development were then compared
with earlier stages for responsiveness to IL-7. Pro-T3 cells were
isolated by sorting from day 16 embryos using CD25 and CD44
markers. As shown in Figure 3, pro-T3 cells were protected by
IL-7 from cell death during culture. Of the first three stages (pro-
T1, -T2, and -T3), the most dramatic IL-7 trophic effects were
consistently observed at pro-T2.
Pro-T4 cells were sorted from day 17 embryos by selecting for
CD4
2
, CD8
2
, CD25
2
, and CD44
2
cells. As shown in Figure 3,
these cells survived independently of IL-7 in culture, and addition
of IL-7 produced only a slight increase in survival that was not
observed consistently. Pro-T4 cells also rapidly differentiated in
culture (not shown), expressing CD4 and CD8 after overnight cul-
ture, and this, too, was unaffected by IL-7. Pro-T4 cells were also
cycling much more rapidly than pro-T1, -T2, and -T3 cells (not
shown), and this cycling was unaffected by IL-7.
FIGURE 1. IL-7 protects day 14 embry-
onic thymocytes from apoptotic death. Thy-
mocytes were cultured with or without IL-7
for varying times. Recovered cells were ana-
lyzed by flow microfluorometry for viability
using exclusion of propidium iodide (left) and
apoptosis using binding of annexin-FITC
(right).
5736 TROPHIC ACTION OF IL-7 ON PRO-T CELLS
After the pro-T4 stage, thymocytes express CD4 and CD8. We
examined CD4
1
CD8
1
cells for IL-7 trophic effects (not shown)
and observed that, like pro-T4 cells, these cells survived indepen-
dently of IL-7 for several days in vitro. We conclude that the
trophic effects of IL-7 end with the pro-T3 stage.
The role of p53 in the apoptotic response to IL-7 deprivation
was tested for two reasons. First, p53 mediates one type of thy-
mocyte apoptosis, that induced by dsDNA breaks (20, 22, 23).
Second, p53
2/2
mice develop thymic lymphomas at a very high
frequency (24, 25), suggesting that they evade the normal death
mechanisms. Although the phenotype of such p53
2/2
thymomas is
primarily CD4
1
CD8
1
, an IL-7-independent stage, we neverthe-
less considered it possible that the actually transformed cell was a
pro-T cell because such thymic lymphomas also arise in rag
2/2
p53
2/2
mice (24) (S. Cande`ias and S. Durum, unpublished obser-
vation) that do not develop beyond the IL-7-dependent pro-T3
stage. However, a role for p53 was ruled out by using pro-T cells
from p53
2/2
mice, as shown in Figure 4. Thus, a similar depen-
dency on IL-7 was observed comparing p53
2/2
thymocytes to
their heterozygous littermates. Note that cell death (in the absence
of IL-7) was not as extensive in this experiment as that shown in
Figure 3; this is because adult pro-T cells die more slowly in cul-
ture than their fetal counterparts. We also tested fetal pro-T cells
from p53
2/2
mice (not shown) and similarly showed that they are
IL-7 dependent. It can be noted (Fig. 4) that p53
2/2
thymocytes
survive better in culture than do their heterozygous counterparts.
This improved survival of cultured p53
2/2
cells was not observed
in studies using unfractionated thymocytes or pre-B cells (20, 22,
23); however, we consistently observed it in pro-T cells and acti-
vated mature T cells (S. Cande`ias and S. Durum, unpublished ob-
servations), and it has been noted in embryonic fibroblasts (26).
A cascade of caspases is a feature of the apoptotic response, and
inhibiting this cascade has been shown to block cell death in many
studies. Caspase inhibitors were added to thymocyte cultures, and
the effect on IL-7 deprivation was studied. Two different inhibitors
were used: z-VAD, which inhibits caspases 1 and 4, and z-DEVD,
which inhibits caspases 3, 6, 7, 8, and 10. As shown in Figure 5
(left panel), neither caspase inhibitor prolonged life, as assessed by
propidium iodide exclusion. However, both caspase inhibitors no-
tably reduced DNA fragmentation (right panel). Thus, IL-7 de-
privation activates caspases (recognized by these inhibitors), lead-
ing to DNA fragmentation; however, an additional mechanism
must also be involved in cell killing when IL-7 is withdrawn; this
mechanism could involve caspases not blocked by these inhibitors
or may be caspase independent.
FIGURE 2. Effect of IL-7 on apoptosis and cell
cycle in stage 1 and stage 2 pro-T cells. Thymocytes
from day 14 embryos were sorted into pro-T1 and
pro-T2 subsets using CD25 and CD44 markers. Cells
were cultured for 24 h with or without IL-7. Recov-
ered cells were analyzed for DNA content.
FIGURE 3. Effect of IL-7 on survival of subsets of pro-T cells. Thy-
mocytes from different days of gestation were sorted into the indicated
populations using CD25 and CD44 markers. Pro-T1 and -T2 cells were
sorted from suspensions of day 14 embryonic thymus, pro-T3 cells were
sorted from day 16 embryonic thymus, and pro-T4 cells were sorted from
day 17 embryonic thymus. Cells were cultured for 24 h with or without
IL-7 and analyzed for viability using exclusion of propidium iodide.
FIGURE 4. Thymocyte death from IL-7 deprivation is not dependent on
p53. Adult thymocytes (from 8-wk-old p53
2/2
or p53
1/2
mice) were
sorted to yield the CD25
1
CD4
2
CD8
2
fraction (pro-T2 and -T3). Cells
were cultured for 24 h and analyzed for viability using exclusion of pro-
pidium iodide.
5737The Journal of Immunology
FasL signals through Fas, inducing apoptotic death in peripheral
T cells, and this pathway also appears to operate in late pro-T4 and
early double-positive cells (27). Two methods were used to test
whether Fas is involved in death from IL-7 deprivation. First, anti-
Fas Ab was added to cultures of thymocytes to determine whether
it blocked cell death in the absence of IL-7. Second, pro-T cells
from MRL-lpr/lpr mice, which lack functional Fas, were also
tested. As shown in Figure 6 (left), anti-Fas did not block the death
of pro-T cells, nor did the MRL-lpr/lpr thymocytes fail to die in
the absence of IL-7. As a positive control for anti-Fas blocking, the
same Ab was shown to inhibit the FasL-Fas killing pathway of
FasL, expressing d11S killers on the Fas-expressing Renca target
cells (Fig. 6, right). Although this Ab can have agonist activity and
kill Fas targets, we do not observe this unless it is secondarily
cross-linked (T. Sayers unpublished observation). Further evi-
dence against a Fas mechanism is our failure to detect Fas expres-
sion by flow microfluorometry before or after culture of pro-T cells
(not shown). Thus, Fas is not involved in the death of pro-thymo-
cytes deprived of IL-7.
The relative concentrations of antiapoptotic (e.g., Bcl-2) and
proapoptotic members (e.g., Bax) of the Bcl-2 family determine
whether a cell will live or die. Intracellular staining was performed
for the antiapoptotic factors Bcl-2 and Bcl-X
L
and for the pro-
apoptotic factors Bax and Bad. As shown in Figure 7 (left), freshly
isolated cells expressed high levels of Bcl-2 and Bax, whereas no
Bcl-X
L
or Bad was detected. During culture (Fig. 7, right panels),
Bcl-2 levels declined sharply, and IL-7 reduced this decline. Bax
levels increased during culture, and IL-7 prevented this increase.
Thus, IL-7 preserved the ratio of Bcl-2 to Bax at a level interme-
diate between that at initiation and that of cultures deprived of
IL-7. Therefore, the trophic action of IL-7 on day 14 thymocytes
correlated with its maintaining a favorable Bcl-2/Bax ratio.
Discussion
We analyzed the effect of IL-7 on survival of pro-T cells. IL-7
protected pro-T1, -T2, and -T3 cells from rapid apoptotic death,
whereas pro-T4 cells survived independently of IL-7. IL-7 did not
induce cell cycling of pro-T cells. There was no evidence for the
involvement of p53 or Fas pathways in cell death. Inhibiting
caspases inhibited DNA fragmentation, but did not protect cells
from death. IL-7 protected pro-T cells from a decline in Bcl-2 and
a rise in Bax, suggesting that the Bcl-2/Bax ratio could be the basis
of the trophic action of IL-7.
These findings complement the recent reports that a bcl-2 trans-
gene restores
ab
T cell development in IL-7R
2/2
mice (28, 29) and
in
g
c
2/2
mice (30), demonstrating that Bcl-2 can substitute for
some IL-7 activities. It has also recently been reported that IL-7
regulates Bcl-2 levels in pro-T cells, and that this regulation is
FIGURE 5. Effects of caspase inhibitors on cell death and DNA fragmentation in the absence of IL-7. Thymocytes from day 14 embryos were cultured
with or without IL-7 and caspase inhibitors. After 24 h, recovered cells were analyzed for viability using exclusion of propidium iodide (left) and DNA
content (right).
FIGURE 6. Thymocyte death from IL-7
deprivation is not dependent on Fas. Thymo-
cytes from day 14 embryos were cultured
with or without IL-7 and anti-Fas Ab, and
cell viability was measured 24 h later (left).
Thymocytes from C57BL/6 mice are com-
pared with the Fas-deficient strain MRL-lpr/
lpr. As a positive control for anti-Fas block-
ade of cell death, D11S cells were mixed
with IFN-treated Renca cells labeled with
51
Cr, with or without anti-Fas, and cell death
was measured 18 h later (right).
5738 TROPHIC ACTION OF IL-7 ON PRO-T CELLS
deficient in IL-7
2/2
mice (31). Taken together with our observa-
tion that IL-7 influences the Bcl-2/Bax ratio in vitro, it suggests
that these regulators of apoptosis could mediate the IL-7 response.
It has previously been shown that IL-7 enhanced the Bcl-2 levels
in cell lines (32, 33), and our study extends this to pro-T cells,
which have a physiologic requirement for IL-7R
a
signals. How-
ever, Bcl-2 induction may not completely explain the trophic ef-
fects of IL-7 on pro-T cells in embryonic life. Mice lacking Bcl-2
have relatively normal numbers of thymocytes at birth; however,
these numbers rapidly decline by 1 mo (34). The decline in thy-
mocytes after birth in these mice has been attributed to differences
in stem cells that seed the thymus before and after birth (35). Thus,
it was proposed that Bcl-2 was not required for embryonic thy-
mopoiesis generated by stem cells derived from fetal liver. At
birth, thymopoiesis is subsumed by a stem cell arriving from bone
marrow, and it or its progeny are dependent on Bcl-2. Perhaps
another Bcl-2 family member is also induced by IL-7 in fetal cells.
We sought two other Bcl-2 family members; Bcl-X
L
protein was
not detected in pro-T cells (Fig. 7), nor was bcl-w (36) mRNA
detectable by Northern blotting (not shown). Remaining family
members that have not been evaluated are Mcl-1 (37) and A1 (38).
The trophic activity of IL-3 on a cell line was attributable to phos-
phorylation of Bad (39); this mechanism would be resistant to
cycloheximide, as the IL-7 trophic effect is in part; however, we
did not detect Bad in pro-T cells (Fig. 7).
Blocking caspase activity is reported to inhibit apoptosis in
many cell types (reviewed in Ref. 40). It is therefore surprising that
caspase inhibitors, although inhibiting DNA fragmentation, did not
preserve the viability of these pro-T cells. Several lines of evidence
suggest that these cells die by apoptosis, including the phenotype
of the cells (Figs. 1 and 2) and the observation that Bcl-2 protects
them (28–30). One possibility is that death is mediated by caspases
that are insensitive to the effects of these inhibitors. Another pos-
sibility is that the death pathway does not involve caspases at all.
This is reminiscent of cytotoxic T cell killing, which is primarily
mediated by perforin; caspase inhibitors block DNA fragmenta-
tion, but not cell death, which in that case is caused by pores in the
plasma membrane (41).
It was observed that wortmannin inhibited the trophic action of
IL-7 on a pre-B cell line, suggesting that phosphatidylinositol 3-ki-
nase is required (42). The same study noted that a tyrosine site on
IL-7R
a
that mediated activation of phosphatidylinositol 3-kinase
was required for the trophic activity in pre-B cells. However, we
did not observe that wortmannin inhibited the trophic activity of
IL-7 on pro-T cells (data not shown). Hence, the seemingly similar
activities of IL-7 on survival of early T and B cells may involve
different signaling pathways and, by extension, different survival
and death pathways.
The Fas pathway did not appear to mediate death from IL-7
deprivation. It has been reported that Fas expression does not begin
until the CD4
1
CD8
1
stage. We have confirmed this and also ob-
served that its expression is not induced during IL-7 deprivation
(not shown). A recent report implicates the Fas system in the death
of cells that fail to productively rearrange their TCR
b
genes (27).
This death presumably occurs in cells at stage pro-T3, which, ac-
cording to our findings, also die when deprived of IL-7. The signal
for a successful
b
gene rearrangement is thought to emanate from
the pre-TCR, which incorporates the successfully produced
b
-chain. Thus, the trophic signals from pre-TCR may be different
from those of IL-7R, and this is substantiated by the failure of a
bcl-2 transgene to rescue T cells in rag
2/2
mice (which cannot
generate
b
gene rearrangements), whereas it protects IL-7R
2/2
thymocytes (28). Consistent with this interpretation, it has been
reported that Bcl-2 cannot protect cells from Fas-mediated killing
(43, 44). We also found no evidence for IL-7 serving as a cofactor
together with the pre-TCR signal, since pro-T4 cells survived in-
dependently of IL-7; pro-T4 cells also rapidly proliferated and dif-
ferentiated into CD4
1
CD8
1
cells within 18 h in the absence of
IL-7 (not shown).
We recently observed that the IL-7R activates the
a
4
b
1
integrin,
increasing its affinity for the extracellular matrix protein fibronec-
tin (45). Integrins, in turn, are known to provide viability signals to
some types of cells. However, we tested whether fibronectin could
augment the trophic effect of IL-7 on pro-T cells and could not
detect such an effect (data not shown). IL-7 produced by thymic
epithelial cells appears to be bound to extracellular matrix in the
thymus (45), which raises questions about the molecular form,
solubility, and concentration of the IL-7 actually encountered by
pro-T cells. Thus, we do not know whether the concentrations of
IL-7 used in our experiments fall into the physiologic or the phar-
macologic range.
We did not observe an induction of cell cycle progression by
IL-7, but, rather, found a completion of the cycle and accumulation
of cells in G
1
. There are reports that IL-7 induced the growth of
pro-T cells (for example, Ref. 46), but these effects may be attrib-
utable to the presence of other stimuli that in some studies were
FIGURE 7. Effect of IL-7 on levels of
Bcl-2 and Bax. Intracellular staining for
Bcl-2, Bcl-X
L
, Bax, and Bad was performed
on thymocytes before and after culture in
IL-7 for the indicated times. Right, Data are
expressed as specific staining, obtained by
subtracting the mean specific fluorescence
(on a linear scale) of the control staining at
each time point.
5739The Journal of Immunology
intentionally added or in other cases may have been produced
endogenously; it is also possible that in long term cultures, a sub-
population of cells that grows in response to IL-7 eventually dom-
inates; however, it can be seen from our results (and inferred from
the bcl-2 transgenic mice noted above (28–30)) that this is not the
prevailing response. Stem cell factor has been identified as a potent
cofactor that, together with IL-7, induces rapid growth (47). Other
cytokines may be produced by the thymocytes during culture in
our studies. The evidence for such autocrine factors is that cultur-
ing the cells at high density in U-bottom wells promoted the tro-
phic effects of IL-7, and conditioned medium from high cell den-
sity cultures promoted the survival of low cell density cultures (not
shown).
Differentiation of thymocytes has also been reported to be in-
duced by IL-7. VDJ recombination is promoted by the IL-7R in
thymocytes (3, 14, 48, 49) and pro-B cells (reviewed in Ref. 1; 42).
This effect is partly attributable to inducing rag1 and rag2 expres-
sion (48, 50, 51) and perhaps also to enhancing locus accessibility,
especially of the TCR
g
locus. We examined whether IL-7 induced
expression of CD25 on pro-T1 cells, since CD25
1
cells are absent
in IL-7R
2/2
mice, but this was not detectable (not shown). How-
ever, we did observe significant induction of CD8
a
and -
b
surface
expression on pro-T cells after overnight culture with IL-7 (K.
Kim, manuscript in preparation).
In conclusion, IL-7 has a trophic effect on several early stages of
pro-T cells, promoting survival without growth. Bcl-2 and Bax
levels are associated with these effects, but there may be other
mechanisms that remain to be identified for this type of biologic
activity.
Acknowledgments
We thank Drs. R. Fenton and J. Oppenheim for comments on the manu-
script, Dr. R. Kirken for wortmannin, L. Finch for flow cytometry, and
R. Wiles for technical assistance.
References
1. Cande`ias, S., K. Muegge, and S. K. Durum. 1997. IL-7 receptor and VDJ re-
combination: trophic versus mechanistic actions. Immunity 6:501.
2. Peschon, J. J., P. J. Morrissey, K. H. Grabstein, F. J. Ramsdell, E. Maraskovsky,
B. C. Gliniak, L. S. Park, S. F. Ziegler, D. E. Williams, C. B. Ware, J. D. Meyer,
and B. L. Davison. 1994. Early lymphocyte expansion is severely impaired in
interleukin 7 receptor-deficient mice. J. Exp. Med. 180:1955.
3. Maki, K., S. Sunaga, and K. Ikuta. 1996. a). The V-J recombination of T cell
receptor-
g
genes is blocked in interleukin-7 receptor-deficient mice. J. Exp. Med.
184:2423.
4. von Freeden-Jeffry, U., P. Vieira, L. A. Lucian, T. McNeil, S. E. Burdach, and
R. Murray. 1995. Lymphopenia in interleukin (IL)-7 gene-deleted mice identifies
IL-7 as a nonredundant cytokine. J. Exp. Med. 181:1519.
5. Peschon, J. J., B. C. Gliniak, P. Morrissey, and E. Maraskovsky. 1997. Lymphoid
development and function in IL-7R deficient mice. In Cytokine Knockouts.
S. K. Durum and K. Muegge, eds. Humana Press, Totowa, NJ, pp. 37–52.
6. Grabstein, K. H., T. J. Waldschmidt, F. D. Finkelman, B. W. Hess, A. R. Alpert,
N. E. Boiani, A. E. Namen, and P. J. Morrissey. 1993. Inhibition of murine B and
T lymphopoiesis in vivo by an anti-interleukin 7 monoclonal antibody. J. Exp.
Med. 178:257.
7. Bhatia, S. K., L. T. Tygrett, K. H. Grabstein, and T. J. Waldschmidt. 1995. The
effect of in vivo IL-7 deprivation on T cell maturation. J. Exp. Med. 181:1399.
8. Noguchi, M., H. Yi, H. M. Rosenblatt, A. H. Filipovich, S. Adelstein,
W. S. Modi, O. W. McBride, and W. J. Leonard. 1993. Interleukin-2 receptor
g
chain mutation results in X-linked severe combined immunodeficiency in hu-
mans. Cell 73:147.
9. DiSanto, J. P., W. Muller, D. Guy-Grand, A. Fischer, and K. Rajewsky. 1995.
Lymphoid development in mice with a targeted deletion of the interleukin 2
receptor
g
chain. Proc. Natl. Acad. Sci. USA 92:377.
10. Cao, X., E. W. Shores, J. Hu-Li, M. R. Anver, B. L. Kelsall, S. M. Russell,
J. Drago, M. Noguchi, A. Grinberg, E. T. Bloom, W. E. Paul, S. I. Katz,
P. E. Love, and W. J. Leonard. 1995. Defective lymphoid development in mice
lacking expression of the common cytokine receptor
g
chain. Immunity 2:223.
11. Maraskovsky, E., M. Teepe, P. J. Morrissey, S. Braddy, R. E. Miller,
D. H. Lynch, and J. J. Peschon. 1996. Impaired survival and proliferation in IL-7
receptor-deficient peripheral T cells. J. Immunol. 157:5315.
12. He, Y. W., and T. R. Malek. 1996. Interleukin-7 receptor
a
is essential for the
development of
gd
1
T cells, but not natural killer cells. J. Exp. Med. 184:289.
13. Maki, K., S. Sunaga, Y. Komagata, Y. Kodaira, A. Mabuchi, H. Karasuyama,
K. Yokomuro, J. Miyazaki, and K. Ikuta. 1996. b). Interleukin 7 receptor-defi-
cient mice lack
gd
T cells. Proc. Natl. Acad. Sci. USA 93:7172.
14. Cande`ias, S., J. J. Peschon, K. Muegge, and S. K. Durum. 1997. Defective T-cell
receptor
g
gene rearrangement in interleukin 7 receptor knockout mice. Immunol.
Lett. 57:9.
15. Perumal, N. B. T. W. Kenniston, D. J. Tweardy, K. F. Dyer, R. Hoffman,
J. Peschon, and P. M. Appasamy. 1997. TCR-gamma genes are rearranged but
not transcribed in IL-7R alpha-deficient mice. J. Immunol. 158:5744.
16. Pribyl, J. A. R., and T. W. LeBien. 1996. Interleukin 7 independent development
of human B cells. Proc. Natl. Acad. Sci. USA 93:10348.
17. Shortman, K., and L. Wu. 1996. Early T lymphocyte progenitors. Annu. Rev.
Immunol. 14:29.
18. Conlon, P. J., P. J. Morrissey, R. P. Nordan, K. H. Grabstein, K. S. Prickett,
S. G. Reed, R. Goodwin, D. Cosman, and A. E. Namen. 1989. Murine thymo-
cytes proliferate in direct response to interleukin-7. Blood 74:1368.
19. Shinkai, Y., S. Koyasu, K. Nakayama, K. M. Murphy, D. Y. Loh, E. L. Reinherz,
and F. W. Alt. 1993. Restoration of T cell development in RAG-2-deficient mice
by functional TCR transgenes. Science 259:822.
20. Lowe, S. W., E. M. Schmittt, S. W. Smith, B. A. Osborne, and T. Jacks. 1993.
p53 is required for radiation-induced apoptosis in mouse thymocytes. Nature
362:847.
21. Telford, W. G., L. E. King, and P. J. Fraker. 1991. Evaluation of glucocorticoid-
induced DNA fragmentation in mouse thymocytes by flow cytometry. Cell Prolif.
24:447.
22. Clarke, A. R., C. A. Purdie, D. J. Harrison, R. G. Morris, C. C. Bird,
M. L. Hooper, and A. H. Wyllie. 1993. Thymocyte apoptosis induced by p53-
dependent and independent pathways. Nature 362:849.
23. Strasser, A., A. W. Harris, T. Jacks, and S. Cory. 1994. DNA damage can induce
apoptosis in proliferating lymphoid cells via p53-independent mechanisms inhib-
itable by bcl-2. Cell 79:329.
24. Mombaerts, P., C. Terhorst, T. Jacks, S. Tonegawa, and J. Sancho. 1995. Char-
acterization of immature thymocyte lines derived from T-cell receptor or recom-
bination activating gene 1 and p53 double mutant mice. Proc. Natl. Acad. Sci.
USA 92:7420.
25. Purdie, C. A., D. J. Harrison, A. Peter., L. Dobbie, S. White, S. E. M. Howie,
D. M. Salter, C. C. Bird, A. H. Wyllie, M. L. Hooper, and A. R. Clarke. 1994.
Tumour incidence, spectrum and ploidy in mice with a large deletion in the p53
gene. Oncogene 9:603.
26. Serrano, M., A. W. Lin, M. E. McCurrach, D. Beach, and S. W. Lowe. 1997.
Oncogenic ras provokes premature cell senescence associated with accumulation
of p53 and p16INK4a. Cell 88:593.
27. Yasutomo, K., K. Maeda, H. Hisaeda, R. A. Good, Y. Kuroda, and K. Himeno.
1997. The fas-deficient SCID mouse exhibits the development of T cells in the
thymus. J. Immunol. 158:4729.
28. Maraskovsky, E., L. A. O’Reilly, M. Teepe, L. M. Corcoran, J. J. Peschon, and
A. Strasser. 1997. Bcl-2 can rescue T lymphocyte development in interleukin-7
receptor deficient mice but not in mutant rag-1
2/2
mice. Cell 89:1011.
29. Akashi, K., M. Kondo, U. von Freeden-Jeffry, R. Murray, and I. L. Weissman.
1997. Bcl-2 rescues T lymphopoiesis in interleukin-7 receptor-deficient mice.
Cell 89:1033.
30. Kondo, M., K. Akashi, J. Domen, K. Sugamura, and I. L. Weissman. 1997. Bcl-2
rescues T lymphopoiesis, but not B or NK cell development in common
g
chain-
deficient mice. Immunity 7:155.
31. von Freeden-Jeffry, U., N. Solvason, M. Howard, and R. Murray. 1997. The
earliest T lineage-committed cells depend on IL-7 for Bcl-2 expression and nor-
mal cell cycle progression. Immunity 7:147.
32. Hernandez-Caselles, T., M. Martinez-Esparza, D. Sancho, D., G. Rubio, and
P. Aparicio. 1995. Interleukin-7 rescues human activated T lymphocytes from
apoptosis induced by glucocorticosteroids and regulates bcl-2 and CD25 expres-
sion. Hum. Immunol. 43:181.
33. Lee, S. H., N. Fumita, T. Mashima, and T. Tsuruo. 1996. Interleukin-7 inhibits
apoptosis of mouse malignant T-lymphoma cells by both suppressing the CPP32-
like protease activation and inducing the Bcl-2 expression. Oncogene 13:2131.
34. Veiss, D. J., C. M. Sorenson, J. R. Shutter, and S. J. Korsmeyer. 1993. Bcl-2-
deficient mice demonstrate fulminant lymphoid apoptosis, polycystic kidneys,
and hypopigmented hair. Cell 75:229.
35. Matsuzaki, Y., K.-I. Nakayama, K. Nakayama, T. Tomita, M. Isoda, D. Y. Loh,
and H. Nakauchi. 1997. Role of bcl-2 in the development of lymphoid cells from
the hematopoietic stem cell. Blood 89:853.
36. Gibson, L., S. P. Holmgreen, D. C. Huang, O. Bernard, N. G. Copeland,
N. A. Jenkins, G. R. Sutherland, E. Baker, J. M. Adams, and S. Cory. 1996.
bcl-w, a novel member of the bcl-2 family, promotes cell survival. Oncogene
13:665.
37. Zhou, P., L. Qian, K. M. Kozopas, and R. W. Craig. 1997. Mcl-1, a Bcl-2 family
member, delays the death of hematopoietic cells under a variety of apoptosis-
inducing conditions. Blood 89:630.
38. Lin, E. Y., A. Orlofsky, M. S. Berger, and M. B. Prystowsky. 1993. Character-
ization of A1, a novel hemopoietic-specific early-response gene with sequence
similarity to bcl-2. J. Immunol. 151:1979.
39. Zha, J., H. Harada, E. Yang, J. Jockel, and S. J. Korsmeyer. 1996. Serine phos-
phorylation of death agonist BAD in response to survival factor results in binding
to 14-3-3 not BCL-X
L
. Cell 87:619.
40. Miller, D. K. 1997. The role of the caspase family of cysteine proteases in ap-
optosis. Semin. Immunol. 9:35.
5740 TROPHIC ACTION OF IL-7 ON PRO-T CELLS
41. Sarin, A., M. S. Williams, M. A. Alexander-Miller, J. A. Berzofsky,
C. M. Zacharchuk, and P. A. Henkart. 1997. Target cell lysis by CTL granule
exocytosis is independent of ICE/Ced-3 family proteases. Immunity 6:209.
42. Corcoran, A. E., F. M. Smart, R. J. Cowling, T. Crompton, M. J. Owen, and
A. R. Venkitaraman. 1996. The interleukin-7 receptor
a
chain transmits distinct
signals for proliferation and differentiation during B lymphopoiesis. EMBO J.
15:1924.
43. Strasser, A., A. W. Harris, D. C. S. Huang, P. H. Krammer, and S. Cory. 1995.
Bcl-2 and Fas/APO-1 regulate distinct pathways to lymphocyte apoptosis. EMBO
J. 14:6136.
44. Moreno, M. B., S. A. Memon, and C. M. Zacharchuk. 1996. Apoptosis signaling
pathways in normal T cells. J. Immunol. 157:3845.
45. Kitazawa, H., K. Muegge, R. Badolato, J. Wang, W. E. Fogler, D. K. Ferris,
C. Lee, S. Candeias, M. R. Smith, J. J. Oppenheim, and S. K. Durum. 1997. IL-7
activates
a
4
b
1
integrin in murine thymocytes. J. Immunol. 159:2259.
46. Moore, T. A., and A. Zlotnik. 1995. T lineage commitment and cytokine re-
sponses of thymic progenitors. Blood 86:1850.
47. Morrissey, P. J., H. McKenna, M. B. Widmer, S. Braddy, R. Voice, K. Charrier,
D. E. Williams, and J. D. Watson. 1994. Steel factor (c-kit ligand) stimulates the
in vitro growth of immature CD3-/CD4-CD8- thymocytes. Cell. Immunol. 157:
118.
48. Muegge, K., M. P. Vila, and S. K. Durum. 1993. Interleukin-7: a cofactor for
V(D)J rearrangement of the T cell receptor
b
gene. Science 261:93.
49. Crompton, T., S. V. Outram, J. Buckland, and J. J. Owen. 1997. A transgenic T
cell receptor restores thymocyte differentiation in interleukin-7 receptor
a
chain-
deficient mice. Eur. J. Immunol. 27:100.
50. Appasamy, P. M., T. W. Kenniston, Y. Weng, E. C. Holt, J. Kost, and
W. H. Chambers. 1993. Interleukin 7-induced expression of specific T cell re-
ceptor
g
variable region genes in murine fetal liver cultures. J. Exp. Med. 178:
2201.
51. Tagoh, H., H. Kishi, A. Okumura, T. Kitagawa, T. Nagata, K. Mori, and
A. Muraguchi. 1996. Induction of recombination activating gene expression in a
human lymphoid progenitor cell line: requirement of two separate signals from
stromal cells and cytokines. Blood 89:4463.
5741The Journal of Immunology
... Binding to IL-7R, IL-7 activates JAK-STAT, PI3K, and Src phosphorylation signaling pathways to regulate target gene expression, including up-regulation of antiapoptotic genes Bcl-2, Bcl-xL and Mcl-1 and down-regulation of pro-apoptotic genes Bax and Bak. This contributes to the survival function of IL-7, with so-called trophic effects on lymphoid progenitors and mature lymphoid cells (76)(77)(78).In mice, intestinal gd T defects induced by IL-7 deficiency are restored by targeted expression of IL-7 in enterocytes (79). The ectopic expression of IL-7 in enterocytes does not restore gd T cells defective in other tissues induced by the IL-7 deficiency, such as the thymus (79). ...
Article
Full-text available
Located in the frontline against the largest population of microbiota, the intestinal mucosa of mammals has evolved to become an effective immune system. γδ T cells, a unique T cell subpopulation, are rare in circulation blood and lymphoid tissues, but rich in the intestinal mucosa, particularly in the epithelium. Via rapid production of cytokines and growth factors, intestinal γδ T cells are key contributors to epithelial homeostasis and immune surveillance of infection. Intriguingly, recent studies have revealed that the intestinal γδ T cells may play novel exciting functions ranging from epithelial plasticity and remodeling in response to carbohydrate diets to the recovery of ischemic stroke. In this review article, we update regulatory molecules newly defined in lymphopoiesis of the intestinal γδ T cells and their novel functions locally in the intestinal mucosa, such as epithelial remodeling, and distantly in pathological setting, e.g., ischemic brain injury repair, psychosocial stress responses, and fracture repair. The challenges and potential revenues in intestinal γδ T cell studies are discussed.
... During the development of T cells, IL-7 up-regulated the expression of anti-apoptotic molecule Bcl-2 and down-regulated the expression of pro-apoptotic proteins Bax and Bad to promote cell survival in previous studies (44,45). Our research had found that IL-7 increased the proportion of S phase of the cell cycle and promote the proliferation of CAR-T cells by inhibiting the expression of CDKN1A. ...
Preprint
Full-text available
CAR-T targeting CD19 have achieved significant effects in the treatment of B-line leukemia and lymphoma. However, the treated patients frequently relapsed and could not achieve complete remission. Therefore, improving the proliferation and cytotoxicity of CAR-T cells, reducing exhaustion and enhancing infiltration capacity are still issues to be solved. The IL-7 has been shown to enhance the memory characteristics of CAR-T cells, but the specific mechanism has yet to be elaborated. miRNAs play an important role in T cell activity. However, whether miRNA is involved in the activation of CAR-T cells by IL-7 has not yet been reported. Our previous study had established the 3rd generation CAR-T cells. The present study further found that IL-7 significantly increased the proliferation of anti-CD19 CAR-T cells, the ratio of CD4 + CAR + cells and the S phase of cell cycle. In vivo study showed that IL-7-stimulated CAR-T possessed stronger tumoricidal efficiency. Further we validated that IL-7 induced CAR-T cells had low expression of CDKN1A and high expression of miRNA-98-5p. Additionally, CDKN1A was associated with miRNA-98-5p. Our results, for the first time, suggested IL-7 could conspicuously enhance the proliferation of CAR-T cells through miRNA-98-5p targeting CDKN1A expression, which should be applied to CAR-T production.
... IL-7 is an essential nonredundant cytokine required for T cell development and survival with a unique impact on T cell metabolism [166][167][168][169][170][171]. The role of IL-7 in promoting Glut1 trafficking and glucose uptake via the STAT5-mediated activation of Akt to support T cell survival has been demonstrated in vitro [172]. ...
Article
Full-text available
Despite rapid advances in the field of immunotherapy, the elimination of established tumors has not been achieved. Many promising new treatments such as adoptive cell therapy (ACT) fall short, primarily due to the loss of T cell effector function or the failure of long-term T cell persistence. With the availability of new tools and advancements in technology, our understanding of metabolic processes has increased enormously in the last decade. Redundancy in metabolic pathways and overlapping targets that could address the plasticity and heterogenous phenotypes of various T cell subsets have illuminated the need for understanding immunometabolism in the context of multiple disease states, including cancer immunology. Herein, we discuss the developing field of T cell immunometabolism and its crucial relevance to improving immunotherapeutic approaches. This in-depth review details the metabolic pathways and preferences of the antitumor immune system and the state of various metabolism-targeting therapeutic approaches.
... иЛ-7 является представителем семейства цитокинов с гамма-цепью, в которое входят иЛ-2, иЛ-4, иЛ-5 и иЛ-21 [29]. иЛ-7 играет ключевую роль в модуляции развития Т-клеток, а также гомеостаза наивных Т-лейкоцитов периферической крови и Т-клеток памяти [14,30,31]. Считается, что основным источником иЛ-7 в семенной жидкости служит предстательная железа, в которой иЛ-7 играет важную роль по обеспечению функционирования Т-клеток [32]. ...
Article
Full-text available
The majority of HIV-1 infections in women transmit through vaginal intercourse, in which virus-containing semen is deposited on the cervico-vaginal mucosa. Semen is more than a mere carrier of HIV-1, since it contains many biological factors, in particular cytokines, that may affect HIV-1 transmission. The concentration of interleukin (IL)-7, one of the most prominent cytokines in semen of healthy individuals, is further increased in semen of HIV-1-infected men. Here, we investigated the potential role of IL-7 in HIV-1 vaginal transmission in an ex vivo system of human cervico-vaginal tissue. We simulated an in vivo situation by depositing HIV-1 on cervico-vaginal tissue in combination with IL-7 at concentrations comparable with those measured in semen of HIV-1-infected individuals. We found that IL-7 significantly enhanced virus replication in ex vivo infected cervico-vaginal tissue. Similarly, we observed an enhancement of HIV-1 replication in lymphoid tissue explants. Analysis of T cells isolated from infected tissues showed that IL-7 reduced CD4+ T cell depletion preventing apoptosis, as shown by the decrease in the number of cells expressing the apoptotic marker APO2.7 and the increase in the expression of the anti-apoptotic protein B-cell lymphoma (Bcl)-2. Also, IL-7 increased the fraction of cycling CD4+ T cells, as evidenced by staining for the nuclear factor Ki-67. High levels of seminal IL-7 in vivo may be relevant to the survival of the founder pool of HIV-1-infected cells in the cervico-vaginal mucosa at the initial stage of infection, promoting local expansion and dissemination of HIV infection.
Article
Interleukin 7 Receptor  Severe Combined Immunodeficiency (IL7R-SCID) is a life-threatening disorder caused by homozygous mutations in the IL7RA gene. Defective IL7R expression in humans hampers T cell precursors proliferation and differentiation during lymphopoiesis resulting in absence of T cells in newborns, who succumb to severe infections and death early after birth. Previous attempts to tackle IL7R-SCID by viral gene therapy have shown that unregulated IL7R expression predisposes to leukaemia, suggesting the application of targeted gene editing to insert a correct copy of the IL7RA gene in its genomic locus and mediate its physiological expression as a more feasible therapeutic approach. To this aim, we have first developed a CRISPR/Cas9-based IL7R-SCID disease modelling system that recapitulates the disease phenotype in primary human T cells and hematopoietic stem and progenitor cells (HSPCs). Then, we have designed a knock-in strategy that targets IL7RA exon 1 and introduces via homology directed repair a corrective, promoterless IL7RA cDNA followed by a reporter cassette through AAV6 transduction. Targeted integration of the corrective cassette in primary T cells restored IL7R expression and rescued functional downstream IL7R signalling. When applied to HSPCs further induced to differentiate into T cells in an Artificial Thymic Organoid system, our gene editing strategy overcame the T cell developmental block observed in IL7R-SCID patients, while promoting full maturation of T cells with physiological and developmentally regulated IL7R expression. Finally, genotoxicity assessment of the CRISPR/Cas9 platform in HSPCs using biased and unbiased technologies confirmed the safety of the strategy, paving the way for a new, efficient, and safe therapeutic option for IL7R-SCID patients.
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.
Chapter
Full-text available
The thymus is an essential organ of the immune system since it is the main site of T-lymphocyteT-lymphocyte development as well as in the regulation of adaptive immunity. The thymusThymus has a highly complex structure that includes the thymic stroma and developing thymocytes. The thymic stroma contains dendritic cells, macrophages, epithelial, mesenchymalMesenchymal and vascular elements. This multicellular environment houses several minor stem cell populations including thymic epithelial progenitor cells/thymic epithelial stem cells, mesenchymal stem cells,Mesenchymal stem cells and lymphoid progenitor/stem cells. The chapter discusses the role of these intrathymic stem cell populations for thymic cell architecture generation and function, and for the thymic regeneration after injuryInjury. Classical transplantation and modern bioengineering approaches and strategies for reconstitution of thymic structure and function using stem cell-based technologies have also been discussed in the chapter in the context of their foreseen perspectives and restrictions for different groups of immunocompromisedImmunocompromised patients. These groups include patients, which were thymectomized during heart surgery, chemotherapy-treated cancer patients, elderly people with age-related thymus involution and impaired thymic function, and in some cases, patients with removed thymoma and thymus-associated autoimmuneAutoimmune diseases. Understanding the role of each thymic stem cell population for supporting the thymus structure and function is a significant factor for choosing a promising thymus reconstitution strategy for these specific groups of patients. In this chapter, the author revises his long-time bench experience of the thymic stem cells in the context of current advancements in this cutting area of research.
Chapter
The kappa opioid receptor (KOR) is expressed on a number of hematopoietic cell populations, based on both protein binding analysis and the detection of kappa opioid receptor gene (Oprk1) transcripts. There are prominent Oprk1 splice variants that are expressed in the mouse and human brain cells and leukocytes. The activation of KOR results in reduced antibody production, an inhibition of phagocytic cell activity, an inhibition of T cell development, alterations in the production of various pro-inflammatory cytokines, chemokines, and the receptors for these mediators. Finally, the activation of KOR also leads to the regulation of receptor functional activity of chemokine receptors through the process of heterologous desensitization. The functional activity of KOR is important for the regulation of inflammatory responses and may provide opportunities for the development of therapeutics for the treatment of inflammatory disease states.
Article
Interleukin (IL)-7 plays an important immunoregulatory role in different types of cells. Therefore, it attracts researcher's attention, but despite the fact, many aspects of its modulatory action, as well as other functionalities, are still poorly understood. The review summarizes current knowledge on the interleukin-7 and its signaling cascade in context of cancer development. Moreover, it provides a cancer-type focused description of the involvement of IL-7 in solid tumors, as well as hematological malignancies.The interleukin has been discovered as a growth factor crucial for the early lymphocyte development and supporting the growth of malignant cells in certain leukemias and lymphomas. Therefore, its targeting has been explored as a treatment modality in hematological malignancies, while the unique ability to expand lymphocyte populations selectively and without hyperinflammation has been used in experimental immunotherapies in patients with lymphopenia. Ever since the early research demonstrated a reduced growth of solid tumors in the presence of IL-7, the interleukin application in boosting up the anticancer immunity has been investigated. However, a growing body of evidence indicative of IL-7 upregulation in carcinomas, facilitating tumor growth and metastasis and aiding drug-resistance, is accumulating. It therefore becomes increasingly apparent that the response to the IL-7 stimulus strongly depends on cell type, their developmental stage, and microenvironmental context. The interleukin exerts its regulatory action mainly through phosphorylation events in JAK/STAT and PI3K/Akt pathways, while the significance of MAPK pathway seems to be limited to solid tumors. Given the unwavering interest in IL-7 application in immunotherapy, a better understanding of interleukin role, source in tumor microenvironment, and signaling pathways, as well as the identification of cells that are likely to respond should be a research priority.
Article
IL-7, a cytokine produced by bone marrow and thymic stroma, is a growth factor for B and T lymphocytes very early in their development. The IL-7R is a heterodimer of an alpha-chain that specifically binds IL-7 and the common gamma-chain, gamma(c), which is also a component of the receptors for IL-2, IL-4, IL-9, and IL-15. IL-7 has also been hypothesized to play a role in the differentiation of gammadelta T cells, which is supported by the recent findings that mice deficient in the alpha-chain of the IL-7R (IL-7R alpha -/-) or IL-7 (IL-7 -/-) have a complete absence of gammadelta T cells, but not alphabeta T cells. We show in this work that Vgamma4 and Vgamma6 TCR genes are rearranged, and sterile Vgamma4 and Vgamma6 TCR-gamma transcripts are expressed in IL-7R alpha -/- thymocytes, but these TCR-gamma genes, and Vgamma5, are not transcribed in thymocytes from IL-7R alpha -/- mice. RAG-1 and RAG-2 genes are transcriptionally active in fetal and adult IL-7R alpha -/- thymocytes. The IL-7-inducible transcription factor, STAT5, is not active in the fetal thymus of IL-7R alpha -/- compared with IL-7R alpha +/+ mice. These data point to a specific role for IL-7/IL-7R signaling in regulating the transcriptional activity, possibly mediated by STAT5, of the rearranged TCR-gamma complex during development of gammadelta T cells, and point to mechanistic differences in the regulation of rearrangement of Vgamma4 and Vgamma6 genes vs Vgamma5.
Article
The earliest steps of intrathymic differentiation recently have been elucidated. It has been reported that both CD4lo (CD44+ CD25- c-kit+ CD3- CD4lo CD8-) and pro-T cells (CD44+ CD25+ c-kit+ CD3- CD4- CD8-, representing the next step in maturation) exhibit germline T-cell receptor beta and gamma loci, suggesting that neither population is exclusively committed to the T-cell lineage. Several groups have shown that CD4lo cells retain the capacity to generate multiple lymphoid lineages in vivo; however, the lineage commitment status of pro-T cells is unknown. To determine when T-cell lineage commitment occurs, we examined the ability of sorted CD4lo and pro-T cells to generate lymphoid lineage cells in vivo or in fetal thymic organ cultures (FTOCs). When intravenously injected into scid mice, CD4lo cells generated both T and B cells, whereas the progeny of pro-T cells contained T cells exclusively. Fetal thymic organ cultures repopulated with CD4lo cells contained both T and natural killer (NK) cells, whereas cultures repopulated with pro-T cells contained T cells almost exclusively. These observations strongly suggest that T-cell lineage commitment occurs during the transition of CD4lo to pro-T cells. Because it is likely that the thymic microenvironment plays a critical role in T-cell commitment, we compared the responses of CD4lo and pro-T cells to various cytokine combinations in vitro, as well as the ability of the cultured cells to repopulate organ cultures. Cytokine combinations that maintained T-cell repopulation potential for both CD4lo and pro-T cells were found. CD4lo cells proliferated best in response to the combination containing interleukin-1 (IL-1), IL-3, IL- 6, IL-7, and stem cell factor (SCF). Unlike CD4lo cells, pro-T cells were much more dependent upon IL-7 for proliferation and FTOC repopulation. However, combinations of cytokines lacking IL-7 were found that maintained the T-cell repopulating potential of pro-T cells, suggesting that, whereas this cytokine is clearly very important for normal pro-T cell function, it is not an absolute necessity during early T-cell expansion and differentiation.
Article
IL-7, a cytokine produced by thymic epithelium, was shown to induce adhesion of murine thymocytes to gelatin-coated membranes. A major binding component of gelatin was identified as fibronectin. IL-7-induced adhesion was observed for all of the major thymocyte subsets, including double-negative, double-positive, and single-positive cells, and specific IL-7R were verified on each subset. Fibronectin binding was mediated via alpha4beta1 integrin (VLA-4), which is expressed at high levels on thymocytes. VLA-4 surface expression was not increased following IL-7 treatment, but was shown to undergo rapid tyrosine phosphorylation on the beta1 subunit. This tyrosine phosphorylation was blocked by genistein, which also blocked IL-7-induced adhesion. IL-7 was detected on the extracellular matrix of the thymus, suggesting that it could promote matrix association through an integrin pathway.
Article
In human tumourigenesis the tumour suppressor gene most commonly affected by mutation, inactivation or allele loss is p53. Loss of p53 function is associated both with failure to maintain a normal diploid status and inability to delete cells by apoptosis following DNA damage. To investigate further the role of p53 we have generated mice carrying a large deletion within the gene. All animals homozygous for this deletion develop spontaneous tumours, predominantly lymphomas, by the age of 6 months. 10% of heterozygotes develop a range of neoplasms, with a lower predisposition towards lymphoma, by 9 months. Both tumour incidence and spectrum in heterozygotes differ from those previously reported in another p53 mutant stock, suggesting either difference in exposure to carcinogens between the two stocks, or a role for modulating genes within different genetic backgrounds. Tumours showed frequent loss of diploid status, and the majority of those arising in heterozygotes showed loss of the wild type allele. These findings are consistent with the concept that p53 acts as a tumour suppressor by preventing the propagation of DNA damage to daughter cells.
Article
Extracellular survival factors alter a cell's susceptibility to apoptosis, often through posttranslational mechanisms. However, no consistent relationship has been established between such survival signals and the BCL-2 family, where the balance of death agonists versus antagonists determines susceptibility. One distant member, BAD, heterodimerizes with BCL-X(L) or BCL-2, neutralizing their protective effect and promoting cell death. In the presence of survival factor IL-3, cells phosphorylated BAD on two serine residues embedded in 14-3-3 consensus binding sites. Only the nonphosphorylated BAD heterodimerized with BCL-X(L) at membrane sites to promote cell death. Phosphorylated BAD was sequestered in the cytosol bound to 14-3-3. Substitution of serine phosphorylation sites further enhanced BAD's death-promoting activity. The rapid phosphorylation of BAD following IL-3 connects a proximal survival signal with the BCL-2 family, modulating this checkpoint for apoptosis.
Article
The effects of interleukin 7 (IL-7) on the growth and differentiation of murine B cell progenitors has been well characterized using in vitro culture methods. We have investigated the role of IL-7 in vivo using a monoclonal antibody that neutralizes IL-7. We find that treatment of mice with this antibody completely inhibits the development of B cell progenitors from the pro-B cell stage forward. We also provide evidence that all peripheral B cells, including those of the B-1 and conventional lineages, are derived from IL-7-dependent precursors. The results are consistent with the rapid turnover of B cell progenitors in the marrow, but a slow turnover of mature B cells in the periphery. In addition to effects on B cell development, anti-IL-7 treatment substantially reduced thymus cellularity, affecting all major thymic subpopulations.
Article
Interleukin (IL)-7 is a potent stimulus for immature T and B cells and, to a lesser extent, mature T cells. We have inactivated the IL-7 gene in the mouse germline by using gene-targeting techniques to further understand the biology of IL-7. Mutant mice were highly lymphopenic in the peripheral blood and lymphoid organs. Bone marrow B lymphopoiesis was blocked at the transition from pro-B to pre-B cells. Thymic cellularity was reduced 20-fold, but retained normal distribution of CD4 and CD8. Splenic T cellularity was reduced 10-fold. Splenic B cells, also reduced in number, showed an abnormal population of immature B cells in adult animals. The remaining splenic populations of lymphocytes showed normal responsiveness to mitogenic stimuli. These data show that proper T and B cell development is dependent on IL-7. The IL-7-deficient mice are the first example of single cytokine-deficient mice that exhibit severe lymphoid abnormalities.
Article
A number of previous studies have suggested a key role for interleukin 7 (IL-7) in the maturation of T lymphocytes. To better assess the function of IL-7 in lymphopoiesis, we have deprived mice of IL-7 in vivo by long-term administration of a neutralizing anti-IL-7 antibody. In a previous report (Grabstein, K. H., T. J. Waldschmidt, F. D. Finkelman, B. W. Hess, A. R. Alpert, N. E. Boiani, A. E. Namen, and P. J. Morrissey. 1993. J. Exp. Med. 178:257-264), we used this system to demonstrate the critical role of IL-7 in B cell maturation. After a brief period of anti-IL-7 treatment, most of the pro-B cells and all of the pre-B and immature B cells were depleted from the bone marrow. In the present report, we have injected anti-IL-7 antibody for periods of up to 12 wk to determine the effect of in vivo IL-7 deprivation on the thymus. The results demonstrate a > 99% reduction in thymic cellularity after extended periods of antibody administration. Examination of thymic CD4- and CD8- defined subsets revealed that, on a proportional basis, the CD4+, CD8+ subset was most depleted, the CD4 and CD8 single positive cells remained essentially unchanged, and the CD4-, CD8- compartment actually increased to approximately 50% of the thymus. Further examination of the double negative thymocytes demonstrated that IL-7 deprivation did, indeed, deplete the CD3-, CD4-, CD8- precursors, with expansion of this subset being interupted at the CD44+, CD25+ stage. The proportional increase in the CD4-, CD8- compartment was found to be due to an accumulation of CD3+, T cell receptor alpha, beta + double negative T cells. Additional analysis revealed that anti-IL-7 treatment suppressed the audition/selection process of T cells, as shown by a significant reduction of single positive cells expressing CD69 and heat stable antigen. Finally, the effects of IL-7 deprivation on the thymus were found to be reversible, with a normal pattern of thymic subsets returning 4 wk after cessation of treatment. The present results thus indicate a central role for IL-7 in the maturation of thymic-derived T cells.
Article
Oncogenic ras can transform most immortal rodent cells to a tumorigenic state. However, transformation of primary cells by ras requires either a cooperating oncogene or the inactivation of tumor suppressors such as p53 or p16. Here we show that expression of oncogenic ras in primary human or rodent cells results in a permanent G1 arrest. The arrest induced by ras is accompanied by accumulation of p53 and p16, and is phenotypically indistinguishable from cellular senescence. Inactivation of either p53 or p16 prevents ras-induced arrest in rodent cells, and E1A achieves a similar effect in human cells. These observations suggest that the onset of cellular senescence does not simply reflect the accumulation of cell divisions, but can be prematurely activated in response to an oncogenic stimulus. Negation of ras-induced senescence may be relevant during multistep tumorigenesis.