on TCR Avidity for Self-Antigens CD8 T Cell Sensory Adaptation

CD8 T Cell Sensory Adaptation Dependent
on TCR Avidity for Self-Antigens
This information is current as
of June 15, 2017.
Maria-Elena Marquez, Wilfried Ellmeier, Vanesa
Sanchez-Guajardo, Antonio A. Freitas, Oreste Acuto and
Vincenzo Di Bartolo
J Immunol 2005; 175:7388-7397; ;
doi: 10.4049/jimmunol.175.11.7388
http://www.jimmunol.org/content/175/11/7388
Subscription
Permissions
Email Alerts
This article cites 41 articles, 18 of which you can access for free at:
http://www.jimmunol.org/content/175/11/7388.full#ref-list-1
Information about subscribing to The Journal of Immunology is online at:
http://jimmunol.org/subscription
Submit copyright permission requests at:
http://www.aai.org/About/Publications/JI/copyright.html
Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/alerts
The Journal of Immunology is published twice each month by
The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright © 2005 by The American Association of
Immunologists All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
Downloaded from http://www.jimmunol.org/ by guest on June 15, 2017
References
The Journal of Immunology
CD8 T Cell Sensory Adaptation Dependent on TCR Avidity
for Self-Antigens1
Maria-Elena Marquez,2* Wilfried Ellmeier,‡ Vanesa Sanchez-Guajardo,† Antonio A. Freitas,†
Oreste Acuto,* and Vincenzo Di Bartolo*
T
he mature TCR repertoire is shaped by positive and negative selection processes resulting from interactions with
self-peptide/MHC (spMHC)3 during thymic differentiation. A repertoire emerges that is sufficiently biased toward selfMHC recognition to ensure T cell survival, but is deprived of those
TCRs whose high avidity for spMHC may lead to cellular activation and autoimmunity (1, 2). However, thymic negative selection
may be leaky, as suggested by the detection in the peripheral T cell
pool of self-reactive cells (2, 3), which are thought to be curbed by
various mechanisms, including deletion, anergy, phenotypic skewing, or regulatory T cells (reviewed in Ref. 4). A conceptual framework to explain how self-tolerance is maintained was put forth in
the tunable activation threshold hypothesis by Grossman and Paul
(5). They proposed that although T cells are activated when TCR
ligation induces intracellular signals of sufficient speed and ampli*Molecular Immunology Unit and †Lymphocyte Population Biology Unit, Department of Immunology, Institut Pasteur, Paris, France; and ‡Institute of Immunology,
Medical University of Vienna, Vienna Competence Center, Vienna, Austria
Received for publication November 30, 2004. Accepted for publication September
27, 2005.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
1
This work was supported by grants from the Institut Pasteur and the Centre National
de la Recherche Scientifique (France). M.E.M. was a recipient of a fellowship from
the Fondo Nacional de Ciencia y Tecnologia (Venezuela). V.S.G. was supported by
the Danish Research Agency (642-02-0016) and the Consejo Nacional de Ciencia y
Technologia (Mexico).
2
Address correspondence and reprint requests to Dr. Vincenzo Di Bartolo, Molecular
Immunology Unit, Institut Pasteur, 25 Rue du Docteur Roux, 75724 Paris, Cedex 15,
France. E-mail address: [email protected]
3
Abbreviations used in this paper: spMHC, self-peptide/MHC; DN, double negative;
DP, double positive; dTg, double Tg; FCM, flow cytometry; HSA, heat-stable Ag
(CD24); LAT, linker for activation of T cell; LCMV, lymphocytic choriomeningitis
virus; LN, lymph node; MFI, mean fluorescence intensity; pTyr, phosphotyrosine; SP,
single positive; Tg, transgenic; VSV, vesicular stomatitis virus; wt, wild type.
Copyright © 2005 by The American Association of Immunologists, Inc.
tude, subthreshold stimuli (e.g., generating slow or low amplitude
TCR signals) may lead to a gradual modification of responsiveness
to subsequent stimuli. For example, autoreactive T cells that escaped central deletion or normally selected T cells facing changes
in spMHC concentration when leaving the thymus may modify the
TCR threshold to levels compatible with survival without activating the T cell (6, 7). This principle may apply to thymocyte maturation (8) during which TCR signaling can indeed be adjusted,
e.g., by CD5 expression (9 –11). At the molecular level, adaptation
may operate by changing levels of expression and/or intrinsic activity of some excitation- and de-excitation-inducing factors, such
as activating and inhibitory receptors and/or kinases and phosphatases (7).
A change in expression of CD5 that negatively regulates signaling through the TCR (9) is the best-known example of TCR
signaling tuning, yet it cannot entirely explain central and peripheral modifications of T cell responsiveness (10 –13). Moreover,
although the effects of spMHC deprivation on T cell responsiveness remain controversial (13–15), it has been shown that different
degrees of adaptive tolerance are induced in CD4 T cells chronically exposed to different amounts of nominal Ag (16). Thus, the
mechanisms tuning activation thresholds in T cells may be quite
diverse and remain ill defined.
To investigate whether mature T cells are endowed with adaptive mechanisms coping with excessive autoreactivity, we sought
to increase the excitability of the TCR signaling machinery by
using a previously characterized gain-of-function mutant of the
protein tyrosine kinase Zap70 (17, 18). Zap70 is crucial for T cell
activation being recruited upon TCR engagement to the CD3-␨
complex (19), where its catalytic activity is augmented by phosphorylation of Tyr493 by the Src-related protein tyrosine kinase
Lck (20). Activated Zap70 phosphorylates the T cell adapter linker
for activation of T cells (LAT) that connects to all major signaling
0022-1767/05/$02.00
Downloaded from http://www.jimmunol.org/ by guest on June 15, 2017
Adaptation of the T cell activation threshold may be one mechanism to control autoreactivity. To investigate its occurrence in vivo,
we engineered a transgenic mouse model with increased TCR-dependent excitability by expressing a Zap70 gain-of-function
mutant (ZAP-YEEI) in postselection CD8 thymocytes and T cells. Increased basal phosphorylation of the Zap70 substrate linker
for activation of T cells was detected in ZAP-YEEI-bearing CD8 T cells. However, these cells were not activated, but had reduced
levels of TCR and CD5. Moreover, they produced lower cytokine amounts and showed faster dephosphorylation of linker for
activation of T cells and ERK upon activation. Normal TCR levels and cytokine production were restored by culturing cells in the
absence of TCR/spMHC interaction, demonstrating dynamic tuning of peripheral T cell responses. The effect of avidity for
self-ligand(s) on this sensory adaptation was studied by expressing ZAP-YEEI in P14 or HY TCR transgenic backgrounds.
Unexpectedly, double-transgenic animals expressed ZAP-YEEI prematurely in double-positive thymocytes, but no overt alteration
of selection processes was observed. Instead, modifications of TCR and CD5 expression due to ZAP-YEEI suggested that signal
tuning occurred during thymic maturation. Importantly, although P14 ⴛ ZAP-YEEI peripheral CD8 T cells were reduced in
number and showed lower Ag-induced cytokine production and limited lymphopenia-driven proliferation, the peripheral survival/
expansion and Ag responsiveness of HY ⴛ ZAP-YEEI cells were enhanced. Our data provide support for central and peripheral
sensory T cell adaptation induced as a function of TCR avidity for self-ligands and signaling level. This may contribute to buffer
excessive autoreactivity while optimizing TCR repertoire usage. The Journal of Immunology, 2005, 175: 7388 –7397.
The Journal of Immunology
Materials and Methods
Animals
C57BL/6 mice were purchased from Charles River Laboratory. C57BL/6
Rag⫺/⫺ mice Tg for the P14 TCR (V␣2; V␤8.1) or the anti-HY TCR
(V␣T3.70; V␤8.2) and B6.CD3⑀⫺/⫺ mice were obtained from the Centre
de Distribution, Typage et Archivage Animal. HYRag⫺/⫺ TCR and
P14Rag⫺/⫺ TCR mice were crossed with wild-type (wt) and ZAPYEEI-Tg mice until obtaining P14Rag⫺/⫺ ⫻ ZAP-YEEI and HYRag⫺/⫺ ⫻
ZAP-YEEI. All these strains were maintained in our specific pathogen-free
animal facilities at the Pasteur Institute. All mice studied were 4 –16 wk
of age.
Generation of Tg mice
The Tg construct bearing the CD8-specific enhancer E8I (i.e., Tg-d in Ref.
25) and the minimal murine CD8 promoter was used to generate ZAPYEEI-Tg mice. The original hCD2 reporter gene was excised by SalI digestion of the plasmid and was replaced by the cDNA encoding for ZAPYEEI (17). The SZ1 and SZ2 founders were generated at the Astra-Zeneca
Transgenic Center and the Service d’Experimentation Animale et Transgénèse, respectively, by injecting the NotI-digested expression cassette in
fertilized C57BL/6 ⫻ CBA eggs. Injected ova were placed in pseudopregnant recipients. Offspring were genotyped by tail biopsy and Southern blot
using the entire 1.9-kb ZAP-YEEI coding sequence as a probe. The Tg
lines were maintained by repeated crossing to the C57BL/6 strains for 10
generations.
Abs and flow cytometry (FCM)
The following mAbs were used for flow cytometry: antiCD8␣-allophycocyanin (53-6.7), anti-TCR␤-allophycocyanin (H57-597),
anti-V␣2-biotin (B20.1), anti-CD4-PE (L3T4/RM4-5), anti-CD69-FITC
(H1.2F3), anti-CD25-FITC (7D4), anti-CD24/heat-stable Ag (HSA)-FITC or
-biotin (M1/69), anti-CD44-FITC (IM781), and anti-CD62L-FITC
(MEL14) from BD Pharmingen; anti-HY TCR-biotin (T3.70), antiCD5-PE (53-7.3), anti-IFN-␥-PE (XMG1.2), anti-IL2-PE (JES6-5H4), antiTNF-␣-PE (MP6-XT22), anti-IL-7Ra-PE (A7R34), and anti-TCR␤-PE
(H57-597) from eBioscience. The anti-vesicular stomatitis virus (VSV)-G
mAb (clone P5D4) (27) was produced as an ascitic fluid and purified by
M. C. Wagner (Flow Cytometry Platform, Institut Pasteur, Paris, France).
It was then coupled with Alexa Fluor 488, using a labeling kit from Molecular Probes. Biotin-coupled Abs were revealed by streptavidin-PerCP.
For intracellular staining, cells were fixed with 2% paraformaldehyde in
PBS; permeabilized with PBS, 1% FCS, and 0.05% saponin; and stained
with the relevant Abs. Analyses were performed with a FACSCalibur flow
cytometer and CellQuest analysis software (BD Biosciences). The following Abs were used for cell stimulation, immunoprecipitation, and immunoblotting: anti-CD3⑀ (145-2C11; produced and purified from culture supernatants by protein A chromatography in our laboratory), anti-Zap70
(clone 29; BD Transduction Laboratories), anti-pTyr mAb (clone 4G10;
Upstate Biotechnology), anti-CD28 (37.51) and anti-␤-tubulin (clone 5H1;
BD Pharmingen), anti-ERK (K-23) and anti-phospho-ERK (clone E-4;
Santa Cruz Biotechnology), and anti-LAT rabbit polyclonal Ab (a gift from
Dr. L. E. Samelson, National Cancer Institute, Bethesda, MD). Secondary
Abs were anti-mouse IgG-HRP (Amersham Biosciences Europe) and
anti-hamster IgG and anti-rabbit-HRP (Jackson ImmunoResearch
Laboratories).
CD8 T cells purification, stimulation, immunoprecipitation, and
immunoblotting
Splenic CD8 T cells were purified by positive selection using CD8␣ (Ly-2)
MicroBeads and an AutoMACS magnetic cell sorter (Miltenyi Biotec),
yielding cell suspensions containing 96 –98% CD8 T cells. For immunoprecipitation and immunoblotting, CD8 T cells were stimulated by incubating them for 20 min on ice with 10 ␮g/ml anti-CD3 mAb (2C11) and
subsequently cross-linking with 10 ␮g/ml anti-hamster IgG at 37°C for the
indicated time. Cells were lysed at 4°C in 1% Nonidet P-40 and 1% n-dodecyl-␤-D-maltoside (Sigma-Aldrich) lysis buffer (18). Immunoprecipitation and immunoblotting were performed as described previously (18).
ECL was detected using a Kodak Image Station 440cf (Eastman Kodak)
and quantified using Kodak 1D analysis software.
Cytokine production
Spleen cells from C57BL/6 mice and TCR-Tg mice expressing ZAP-YEEI
were incubated in 96-well plates (2.5 ⫻ 105 cells/well) at 37°C in 5% CO2
in complete RPMI 1640 medium (containing 10% FCS, 5 ⫻ 10⫺5 M
2-ME, 1 mM HEPES, and antibiotics). Cells were stimulated with platebound anti-CD3 and soluble anti-CD28 Abs or by adding the TCR-Tgspecific peptides gp33– 41 and Smcy-3 (28) in the presence of 10 ␮g/ml
brefeldin A (Sigma-Aldrich). After 20 h, cells were collected and stained
for surface Ags. Samples were then fixed, permeabilized, and stained with
IFN-␥-PE, IL-2-PE, or TNF-␣-PE Abs, followed by anti-VSV-Alexa488.
For some experiments, C57BL/6 splenocytes were activated with 10 ng/ml
PMA (Sigma-Aldrich) and 500 ng/ml Ca2⫹ ionophore A23187 (SigmaAldrich) for 6 h before intracellular staining. TCR expression and cytokine
production after in vitro culture was monitored as follows. Purified CD8 T
cells from C57BL/6 mice expressing ZAP-YEEI were cultured in 96-well
plates (2.5 ⫻ 105 cells/well) in complete RPMI 1640 medium supplemented with murine IL-7 (1/100 dilution of a supernatant from J558 cells
transfected with an IL-7 cDNA; a gift from F. Melchers, University of
Basel, Basel, Switzerland) (29) and 10 ␮g/ml of either anti-mouse H-2Db
(28-14-8) or isotype-matched control Ab (G155-178; both from BD
Pharmingen). At the indicated times, cells were stained with anti-CD8allophycocyanin, anti-TCR␤-PE Abs, and anti-VSV-Alexa 488 and analyzed by FCM. Alternatively, cells were stimulated with anti-CD3 and
anti-CD28 immediately or after 72 h of culture, followed after 20 h by
intracellular staining for IFN-␥-PE and anti-VSV-Alexa 488 and FCM.
Peripheral T cell transfers
Nonirradiated male B6.CD3⑀⫺/⫺ hosts were injected i.v. with CD8 lymph
node (LN) T cell populations from P14Rag⫺/⫺ or P14Rag⫺/⫺ ⫻ ZAPYEEI double-Tg (dTg) mice. After removing spleens from hosts at different times after injection, cell suspensions were prepared, and the number
and percentage of cells from each donor population were evaluated. The
origin of the cells (ZAP-YEEI⫹ or ZAP-YEEI⫺ donor) was assessed by
intracellular staining with the anti-VSV mAb.
Results
Generation of Tg mice expressing a Zap70 gain-of-function
mutant in mature CD8 thymocytes and peripheral CD8 T cells
Mouse oocytes were injected with a construct containing the
CD8␣ gene promoter flanked by the E8I enhancer (25), driving
expression of the gain-of-function mutant of Zap70, ZAP-YEEI,
with a C-terminal-appended VSV epitope tag (17) (Fig. 1A). Normally, E8I directs gene expression only in postselected CD8 SP
Downloaded from http://www.jimmunol.org/ by guest on June 15, 2017
pathways (21). We and others have reported that T cell activation
and thymic differentiation also crucially depend on the phosphorylation of tyrosine Tyr319 in interdomain B of Zap70 (18, 22, 23).
Our previous studies demonstrated that phosphorylated Tyr319
generates a site (pY319SDP motif) where Lck binds via its Src
homology 2 domain, thus favoring Zap70 activation (17). Consistently, mutation of Y319SDP to Y319EEI, which augmented LckSrc homology 2 binding affinity for Zap70 (24), leads to a gainof-function of Zap70 (ZAP-YEEI), as detected by the strong
increase in TCR-dependent signaling (17). Based on these data, we
have engineered a transgenic (Tg) mouse model in which ZAPYEEI is expressed in postselection CD8 single-positive (SP) thymocytes and mature CD8 T cells using the CD8-specific enhancer
E8I (also named E8SP) (25, 26). This system allowed us to show
that naive T cells and postselection SP thymocytes dynamically
modify their signaling capacity when confronted with augmented
levels of stimuli by spMHC. Evidence for signaling adaptation in
mature T cells as a function of TCR-spMHC strength of interaction
was obtained when these mice were crossed onto TCR-Tg mice
P14 and HY, representing prototypes of high and low avidity for
spMHC, respectively. Moreover, unexpected premature expression of ZAP-YEEI in double-positive (DP) thymocytes of P14 and
HY TCR-Tg mice allowed us to demonstrate that modulation of
TCR and CD5 expression can occur in response to augmented
spMHC-derived stimuli during thymocyte maturation, possibly
constituting a signaling adaptation mechanism integrating classic
thymic selection schemes. Collectively, our data strongly suggest
that CD8 thymocytes and T cells are able to reprogram their signaling machinery in reaction to TCR-received stimuli that reflect
avidity for spMHC.
7389
7390
TCR AVIDITY-DEPENDENT ADAPTATION OF CD8 T CELLS
DP thymocytes, but it was expressed in CD8 SP cells (Fig. 1C).
The lower frequency of cells expressing ZAP-YEEI in CD8 SP
thymocytes compared with LN CD8 T cells (i.e., 50 vs 75%) was
probably due to maturation-dependent increased expression from
the E8I enhancer (25). Consistently, the amount of ZAP-YEEI in
CD8 SP thymocytes augmented with HSA down-regulation (Fig.
1C), coincident with completion of positive selection (30). ZAPYEEI expression did not alter the frequency of any thymic subset
(percentages for Tg vs wt mice, respectively, were 6.7 ⫾ 0.2 vs
5.1 ⫾ 1.3 for DN, 80.4 ⫾ 1.4 vs 81.8 ⫾ 3.6 for DP, 9.1 ⫾ 2.4 vs
9.6 ⫾ 3.7 for SP CD4, and 3.8 ⫾ 0.9 vs 3.6 ⫾ 1.1 for SP CD8; n ⫽
3 for all data), in particular, the percentage of mature HSAlow CD8
SP thymocytes (Fig. 1C), indicating no overt effect on thymic selection processes. Spleen and LN from ZAP-YEEI-Tg mice also
had normal cellularity (not shown).
ZAP-YEEI-bearing CD8 T cells display reduced levels of TCR
and CD5
thymocytes and mature CD8 T cells (25). Two Tg mouse lines
from independent founders, referred to as SZ1 and SZ2, expressed
similar amounts of ZAP-YEEI protein in LN CD8 T cells, as detected by FCM with anti-VSV mAb (Fig. 1B). However, ZAPYEEI was expressed in 75% SZ1 and 15% SZ2 CD8 T cells,
respectively (Fig. 1B), a variegation expected with E8I-based constructs (25). Because SZ1 and SZ2 shared essential phenotypic
traits, the data reported below refer to the former line, unless otherwise indicated. The amount of Zap70 in transgene-expressing
cells (i.e., including endogenous Zap70 and ZAP-YEEI) was 2- to
3-fold higher than that in control CD8 T cells, as measured both
after immunoprecipitation (Fig. 4A) and in whole cell lysates (not
shown). ZAP-YEEI was undetectable in peripheral CD4 T cells, B
cells, or NK cells (not shown) as well as in CD4 SP or CD4/CD8
FIGURE 2. TCR and CD5 expression in ZAP-YEEIexpressing CD8 T lymphocytes. FCM analysis of cells
stained with anti-CD8-allophycocyanin, anti-CD4-PE,
and anti-TCR␤-PerCP and intracellularly stained with
anti-VSV-Alexa488. A, ZAP-YEEI vs TCR␤ profiles
are displayed for CD8 SP thymocytes and CD8 T lymph
node cells from SZ1 and SZ2 mice. The percentage of
transgene-positive and transgene-negative cells is indicated. B, Expression of CD5 on peripheral CD8 T cells
from ZAP-YEEI Tg (black line) and control mice (u).
Reduced cytokine production by CD8 T cells expressing
ZAP-YEEI
These results prompted examination of the functional response of
CD8 T cells expressing ZAP-YEEI to TCR and CD28 costimulation. As shown in Fig. 3A, IFN-␥ production, as detected by intracellular staining, was appreciably reduced in the ZAP-YEEI⫹
Downloaded from http://www.jimmunol.org/ by guest on June 15, 2017
FIGURE 1. ZAP-YEEI expression in thymocytes and peripheral CD8 T
lymphocytes from transgenic mice. A, Schematic representation of the
transgenic construct (modified from Ref. 25). P8␣, minimal CD8␣ promoter; CD4, splicing module composed of the untranslated exon I, part of
intron I, and part of exon II from the murine CD4 gene; pA, SV40 polyadenylation site. B, Lymph node cells from SZ1 (left panel) and SZ2 (right
panel)-Tg mice, were surface stained with anti-CD8 Abs, then fixed, permeabilized, and stained with the anti-VSV Ab to detect ZAP-YEEI. The
percentage of ZAP-YEEI-positive cells in the CD8 population from ZAPYEEI Tg mice (black line) is indicated within each panel. , Anti-VSV
staining of control cells from wild-type littermates. C, Thymocytes were
analyzed by FCM after staining with anti-CD8-allophycocyanin, anti-CD4PE, anti-HSA-PerCP, and anti-VSV-Alexa488. CD4 vs CD8 dot plots are
displayed for total thymocytes (left panels). Histograms show the expression of ZAP-YEEI (middle panels) and HSA (right panels) in ZAP-YEEI
Tg mice (black line) and wt mice (u) in the thymocyte population gated in
the corresponding left panel.
Spleen and LN CD8 T cells from ZAP-YEEI-Tg mice displayed an
apparent naive phenotype, because they expressed levels of CD8,
CD28, CD69, CD44, CD45, CD25, and CD62L comparable to
those found in C57BL/6 wt littermates and did not up-regulate
CTLA4 (data not shown). Notably, however, ZAP-YEEI⫹-CD8 T
cells from both SZ1 and SZ2 LN showed decreased TCR amounts
(⬃50% reduction in mean fluorescence intensity (MFI) of TCR␤;
Fig. 2A, middle and right panels). A similar decrease in TCR expression was observed in ZAP-YEEI⫹-CD8 T cells from spleen
(not shown) and ZAP-YEEI⫹-CD8 SP thymocytes (Fig. 2A, left
panel). It is unlikely that lower TCR levels were consequent to
deletion of TCRhigh thymocytes, because negative selection was
not noticeably augmented in these animals (see above). The TCR
may be down-regulated as a consequence of ligand binding, followed by internalization and degradation (31). Therefore, it is
likely that stronger signaling after interactions with spMHC may
induce TCR down-regulation in ZAP-YEEI⫹ cells without inducing activation.
CD8 T cells from ZAP-YEEI-Tg mice also displayed a slight,
but reproducible, reduction in CD5 expression (Fig. 2B). CD5 attenuates TCR signaling (9), and its expression level positively correlates with the strength of TCR-induced signals (10, 13). Although the reason for the decrease in CD5 expression in this
setting was unclear (see Discussion), these data suggested that persistent augmentation of spMHC-generated TCR signals in vivo
modulates both TCR and CD5 expression in mature T cells.
The Journal of Immunology
compared with the ZAP-YEEI⫺ subset. Similar results were obtained for TNF-␣ and IL-2 production (Fig. 3C). Both the frequency of cytokine-producing cells and the amount of cytokine
produced per cell were lower (Fig. 3, A and C). Lower IFN-␥ and
IL-2 secretion by CD8 T cells from SZ1 mice was confirmed by
ELISA (not shown). However, cells stimulated with PMA and a
Ca2⫹ ionophore produced equal levels of IFN-␥ (Fig. 3B) and
other cytokines (data not shown) regardless of ZAP-YEEI expression. Thus, expression of ZAP-YEEI decreased activation-induced
cytokine production, a defect apparently caused by attenuation of
TCR-proximal signaling.
FIGURE 4. Biochemical analysis of TCR-proximal signaling in CD8 T
cells from ZAP-YEEI-Tg mice. Lysates from 5 ⫻ 106 purified CD8 T cells
of ZAP-YEEI Tg and wt mice, unstimulated or stimulated for the indicated
time with the anti-CD3 mAb, were immunoprecipitated with the antiZap70 antiserum (A) and analyzed by anti-pTyr immunoblotting (upper
panel). The same blot was reprobed with anti-Zap70. Blots were revealed
by ECL, and images were acquired using a Kodak Image Station CCD
camera (lower panel). B, Phosphorylation of cellular proteins from ZAPYEEI Tg and wt lymphocytes. Total lysates from 1 ⫻ 106 purified CD8 T
cells, unstimulated or stimulated for the indicated time with anti-CD3
mAb, were immunoblotted with an anti-pTyr mAb (upper panel). Protein
loading in each lane was assessed by reprobing with an anti-tubulin Ab
(second panel from top). After acquiring images as described in A, pp36
and tubulin band intensities were quantified. Numbers below the panel
represent relative pp36 intensity normalized by the amount of tubulin in the
same lane. The blot was sequentially stripped and reprobed with anti-pERK
and anti-ERK Abs (third and fourth panels from top). Numbers below the
lower panel represent the relative pERK-2 intensities after normalization
for total ERK-2 in the same sample. A representative experiment of four is
shown. C, Cells treated for the indicated time as described in A were immunoprecipitated with an anti-LAT antiserum, then sequentially analyzed
by anti-pTyr (upper panel) and anti-LAT (lower panel) immunoblotting.
Numbers below the lower panel indicate the relative intensity of pLAT
after normalization for the total LAT content.
Modified TCR-proximal signaling in CD8 T cells from
ZAP-YEEI-Tg mice
To investigate alterations in TCR-proximal biochemical events
that might explain the reduced activation of ZAP-YEEI⫹ cells, the
phosphorylation status of Zap70 in splenic CD8 T cells from normal C57BL/6 and SZ1 mice was examined. Zap70 tyrosine phosphorylation in unstimulated control cells (Zap70wt) was hardly
detectable, but underwent a sharp increase upon anti-CD3 crosslinking (Fig. 4A). In contrast, phosphorylation of ZAP-YEEI (migrating with reduced mobility because of the appended epitope tag;
Fig. 4A, lower panel) was detected even in unstimulated cells, and
its intensity increased upon anti-CD3 stimulation well above the
levels seen in endogenous Zap70wt cells (Fig. 4A, upper panel).
As previously demonstrated in Jurkat cells (17), the higher phosphorylation of ZAP-YEEI compared with Zap70wt correlates with
the higher TCR-induced kinase activity of the former. Consis-
tently, unstimulated cells from SZ1 mice had increased basal phosphorylation of the 36-kDa transmembrane adaptor LAT (3- to
4-fold higher than in normal mice; Fig. 4, B and C), a major Zap70
substrate (21). The hyperphosphorylation of ZAP-YEEI and LAT
was not induced by CD8 cross-linking during the positive selection
procedure we routinely used to purify CD8 T cells, because identical results were obtained with cells isolated by negative selection
(not shown). The magnitude of gain-of-function of ZAP-YEEI previously measured in Jurkat cells (e.g., 20- to 30-fold greater capacity to stimulate NF-AT activation compared with Zap70wt)
(17) makes it unlikely that changes in basal and induced phosphorylation were due to the mere 2- to 3-fold increase in total
Zap70 (endogenous Zap70 plus ZAP-YEEI), although we cannot
Downloaded from http://www.jimmunol.org/ by guest on June 15, 2017
FIGURE 3. Activation-induced cytokine production by ZAP-YEEI-expressing CD8 T cells. Splenocytes (5 ⫻ 105) from wt mice and ZAP-YEEI
Tg mice were incubated with anti-CD3 (10 ␮g/ml) and anti-CD28 (10
␮g/ml) for 20 h (A) or with PMA (10 ng/ml) and Ca2⫹ ionophore (500
ng/ml) for 6 h (B). Cells were then stained for CD8-allophycocyanin expression, fixed, permeabilized, and stained for IFN-␥-PE and anti-VSVAlexa488. The IFN-␥ vs ZAP-YEEI two-color profiles are displayed on
CD8 T cells. The MFIs of cytokine-producing cells are indicated in the dot
plots. Similar results were obtained in at least five other independent experiments. C, Splenocytes from ZAP-YEEI Tg mice were stimulated with
anti-CD3 and anti-CD28 Abs, as explained above, and then stained with
anti-CD8-allophycocyanin, anti-cytokine-PE, and anti-VSV-Alexa488. Intracellular cytokines were measured in ZAP-YEEI-negative (䡺) and ZAPYEEI-positive (f) CD8 cells. For each population, the percentage (left
panel) and the MFI (right panel) of cytokine-producing cells are shown.
7391
7392
rule out a contribution due to such a limited protein overexpression. Thus, although these data agree with the prediction that ZAPYEEI confers higher TCR-proximal signaling, its expression in
peripheral T cells reduced T cell activation. One explanation may
reside in reduced TCR levels and/or other negative feedback
mechanisms compensating for augmented Zap70 function as part
of an adaptive behavior. Indeed, comparison of the kinetics of
anti-CD3-induced phosphorylation in CD8 T cells from SZ1 and
control mice revealed that although after 30-s stimulation with
anti-CD3, LAT phosphorylation was slightly higher in the former,
at later time points this difference disappeared (i.e., 2 min; Fig. 4B)
and then reversed (i.e., 5 and 10 min; Fig. 4, B and C). ERK2 also
underwent faster dephosphorylation kinetics in SZ1 compared
with normal mice (Fig. 4B, lower panels). Collectively, these data
indicate that expression of ZAP-YEEI results in higher basal and
early CD3-induced LAT phosphorylation. However, the activation
signal is down-regulated more quickly, providing a potential explanation for the reduced TCR-induced cytokine production,
which requires sustained signaling (32).
To directly address whether the reduction of TCR levels and cytokine production in ZAP-YEEI-expressing cells was the result of
dynamic signal tuning upon increased self-stimulation, we cultured
purified CD8 T cells from SZ1 mice in the presence of an anti-H2Db Ab to prevent TCR interaction with spMHC. IL-7 was also
added to sustain naive T cell survival in vitro (33). TCR expression
by ZAP-YEEI⫹ cells significantly increased within the first 24 – 48
h. Consequently, the ratio between TCR␤ MFI on ZAP-YEEI⫺
and ZAP-YEEI⫹ CD8 T cells changed from 2 to 1.2 and remained
stable for an additional 48 h (Fig. 5A). The recovery of TCR expression in ZAP-YEEI⫹ cells was accompanied by a functional
resetting of the signaling machinery. Indeed, when we measured
FIGURE 5. TCR expression and cytokine production after in vitro culture of ZAP-YEEI-expressing T cells. Purified CD8 T cells (2.5 ⫻ 105)
from ZAP-YEEI-Tg mice were incubated with anti-H-2Db Abs in the presence of IL-7. A, At the indicated time, cells were triple stained for CD8,
TCR␤, and ZAP-YEEI. Each point represents the ratio of TCR␤ MFI from
ZAP-YEEI-negative and ZAP-YEEI-positive cells (mean ⫾ SD; n ⫽ 4
mice). B, Cells were stimulated with anti-CD3 and anti-CD28 Abs immediately (right panel) or after 72 h of culture with anti-H-2Db (left panel).
Cells were collected 20 h later and stained with anti-CD8, anti-IFN-␥, and
anti-VSV Abs. IFN-␥ was measured in CD8-gated ZAP-YEEI-negative
and ZAP-YEEI-positive cells. For each population, the percentage of cytokine-producing cells is indicated. Similar results were obtained in four
other independent experiments.
CD3/CD28-induced IFN-␥ production by cells cultured for 72 h
under the same conditions, we found that ZAP-YEEI⫹ cells produced the same or even higher levels of IFN-␥ as ZAP-YEEI⫺
cells (20.3 ⫾ 4.7 vs 15.9 ⫾ 3.6; n ⫽ 4; Fig. 5B). These data
indicated that reduced TCR expression and activation in ZAPYEEI-expressing cells required continuous interaction of the TCR
with spMHC.
ZAP-YEEI in TCR-Tg background mice: unpredicted expression
in DP thymocytes and inverse correlation between TCR levels
and avidity for spMHC
The above data showed that TCR-dependent signals induce a phenotypic and functional tuning of mature T cells. One prediction
would then be that TCR avidity for spMHC and negative tuning of
the signaling machinery are directly correlated and that T cells
with the highest down-modulated TCR within the ZAP-YEEI⫹
cells (Fig. 2A) would be those bearing TCR with higher affinity/
avidity for spMHC. To directly test these ideas, SZ1-Tg mice were
crossed onto mouse lines expressing the P14 and HY ␣␤ TCR
transgenes (34, 35). Both these receptors are positively selected by
H-2Db, but P14 displays a higher avidity for spMHC than HY (10,
11, 36). However, one unexpected finding apparently complicated
the study in these dTg mice in that, in contrast to C57BL/6 mice
(Fig. 1), ZAP-YEEI expression started at the DP stage on both
TCR-Tg backgrounds (Fig. 6, A and B). The reason for the earlier
activation of the E8I enhancer is unclear, but it is probably due to
premature expression and signaling of the transgenic ␣␤ TCR in
developing thymocytes (28, 37). Hence, in these models, ZAPYEEI might modify signaling during thymocyte maturation, thus
affecting positive and/or negative selection processes.
FACS analysis of P14 ⫻ ZAP-YEEI dTg thymi revealed ZAPYEEI expression in 40% of DP cells, increasing up to 78% in the
CD8 SP subset (Fig. 6A, middle panels). Similar to SZ1 and SZ2
mice (see Fig. 2), there was an inverse correlation between the
expression of ZAP-YEEI and the P14 TCR (Fig. 6A). In particular,
the V␣2high population, representing cells that up-regulated the
TCR level after positive selection (30), was strongly reduced in
CD8 SP cells and in the CD4int/lowCD8high transitional population
from P14 ⫻ ZAP-YEEI thymi (Fig. 6A, right panels in the last two
rows). A weak TCR down-regulation was also observed in DP
cells (note the slight drift to the right and the increased cell number
of the DP V␣2 lower expressors; Fig. 6A). The expression of CD5
was also weakly reduced in CD8 SP cells (Fig. 6C). Although
reduction of TCRhigh SP thymocytes is normally interpreted as a
sign of increased negative selection, this did not appear to be the
case, because we found that the fraction of mature CD8 SP thymocytes with down-regulated HSA was similar in P14 and P14 ⫻
ZAP-YEEI (Fig. 6C), and no significant differences in the absolute
numbers of CD8 SP thymocytes were detected (Table I). These
data are consistent with maturing P14 thymocytes reacting to a
lower activation threshold conferred by ZAP-YEEI by tuning TCR
levels, hence limiting/avoiding negative selection.
In HY ⫻ ZAP-YEEI female mouse thymi, the expression of
ZAP-YEEI also began at the DP stage (⬃25% of ZAP-YEEI⫹
cells; Fig. 6B) and gradually increased with maturation, reaching
almost 95% in the CD8 SP subset (Fig. 6B). However, in contrast
to P14 ⫻ ZAP-YEEI mice, HY TCR was not down-regulated.
Indeed, T3.70high cells appeared earlier in HY ⫻ ZAP-YEEI than
in control HY mice, concomitant with reduced DP TCRlow cells
(Fig. 6B, right panel in the first row) and were of comparable
frequency in mature CD8 SP thymocytes (Fig. 6B, right panel in
the bottom row). Moreover, the percentage of HSAlow cells (Fig.
6C) and the absolute number of CD8 SP thymocytes (Table I) were
Downloaded from http://www.jimmunol.org/ by guest on June 15, 2017
Recovery of TCR expression and cytokine production in ZAPYEEI-expressing CD8 T cells upon self-stimuli deprivation
TCR AVIDITY-DEPENDENT ADAPTATION OF CD8 T CELLS
The Journal of Immunology
not different from those in HY female littermates. These data indicate that the expression of ZAP-YEEI in DP thymocytes does
not grossly alter their development. Rather, thymocytes appear to
cope with signal excess in a different way, probably dependent on
their high and low avidities for their selecting ligand(s), respectively: down-regulating TCR levels and limiting negative selection
for P14 or accelerating maturation for HY.
Divergent consequences of ZAP-YEEI expression on homeostasis
of peripheral CD8 T cells in P14 and HY transgenic mice
Similar to SZ1 and SZ2 mice, peripheral T cells from P14 ⫻ ZAPYEEI and female HY ⫻ ZAP-YEEI mice expressed higher levels
of ZAP-YEEI than thymocytes (Fig. 7, top panels), and almost all
CD8 T cells (90 –95%) expressed the mutant. No changes in the
expression levels of CD25, CD44, CD62L, and CD69 were evident
in unstimulated cells (data not shown). However, TCR expression
was differently affected in these mice. V␣2⫹ Tg TCR in CD8 T
cells of P14 ⫻ ZAP-YEEI mice was ⬃3.5-fold lower than that in
control P14 littermates (Fig. 7, middle row, left panel). A smaller
reduction (1.7-fold) in clonotypic T3.70 TCR expression was also
seen in HY ⫻ ZAP-YEEI females compared with HY littermates
(Fig. 7, middle right panel). The apparent decrease in either V␣2
or T3.70 staining was not due to the expression of a second TCR
␣-chain induced by stronger signaling, because a similar reduction
in clonotypic TCRs was observed on a Rag⫺/⫺ background, where
the expression of alternative transcripts is suppressed (data not
shown). Similar to SZ1 mice (Fig. 2), CD5 was slightly decreased
in P14 ⫻ ZAP-YEEI CD8 T cells compared with P14 littermates
(Fig. 7, bottom left panel). Surprisingly, CD5 in CD8 T cells from
female HY ⫻ ZAP-YEEI mice was significantly augmented compared with that in control cells (Fig. 7, bottom right panel). Based
on the proposed correlation between CD5 levels and the strength
of TCR-mediated signaling (10, 13), these results suggest that
TCR-dependent signals are reduced in P14 ⫻ ZAP-YEEI T cells,
although they are increased in HY ⫻ ZAP-YEEI T cells. Accordingly, we found that the number of LN CD8 T cells in P14 ⫻
ZAP-YEEI was reduced compared with that in P14 mice, whereas
it was augmented in HY ⫻ ZAP-YEEI mice compared with that in
HY littermates (Table I). Additional confirmation of the reduced
response to spMHC-induced stimuli of P14 ⫻ ZAP-YEEI cells
was obtained by transferring these cells into lymphopenic hosts.
Equal numbers of CD8 T cells purified from P14Rag⫺/⫺ ⫻ ZAPYEEI and P14Rag⫺/⫺ animals were mixed and transferred into
CD3⑀⫺/⫺ syngenic hosts. Spleens were removed at different times
after transfer, and the absolute number and percentage of
P14Rag⫺/⫺ ⫻ ZAP-YEEI- and P14Rag⫺/⫺-derived cells were assessed by intracellular staining with anti-VSV Ab. Although the
number of P14Rag⫺/⫺-derived cells significantly increased after
transfer, a much lower increase was observed for P14Rag⫺/⫺ ⫻
ZAP-YEEI cells (Fig. 8A), demonstrating that the ability of the
latter to undergo lymphopenia-driven expansion was impaired. Interestingly, we observed a slight reduction in IL-7R ␣-chain levels
on P14 cells expressing ZAP-YEEI compared with normal P14 T
cells (Fig. 8B). Although the significance of this finding is still
unclear, IL-7 has been proposed as a major regulator of both survival of naive T cells in normal mice and their expansion in lymphopenic hosts (reviewed in Ref. 33). Thus, decreased IL-7R␣
levels may contribute to the defects observed in P14 ⫻ ZAP-YEEI
cells.
Overall, the expression of ZAP-YEEI in P14- and HY TCRbearing cells suggests that signal threshold tuning is initiated during thymocyte development and could continue in the peripheral
compartment, as suggested by TCR and CD5 level modulation.
Contrasting functional responses in vitro of P14 and HY T cells
expressing ZAP-YEEI
The consequences of the divergent adaptation of CD8 T cells from
dTg mice on the induction of effector functions were assessed by
analyzing their response to Ag stimulation in vitro. Splenocytes
from P14 and P14 ⫻ ZAP-YEEI mice or from female HY and
HY ⫻ ZAP-YEEI mice were cultured in the presence of specific
agonist peptides for 20 h, and cytokine production by CD8 T cells
was determined by intracellular staining and flow cytometry. The
percentage of cells producing IFN-␥, IL-2, and TNF-␣ in response
to saturating concentrations of the antigenic peptide was lower in
P14 ⫻ ZAP-YEEI splenocytes compared with control cells (Fig.
9A), a result reminiscent of that obtained for ZAP-YEEI-expressing T cells from C57BL/6 mice. In contrast to the P14 model, the
Downloaded from http://www.jimmunol.org/ by guest on June 15, 2017
FIGURE 6. Thymic development in P14 and H-Y mice expressing
ZAP-YEEI. FCM analysis of cells stained with anti-clonotypic Abs conjugated to biotin-streptavidin-PerCP, anti-CD8-allophycocyanin, and antiCD4-PE and intracellularly stained for ZAP-YEEI-Alexa488. CD4 vs CD8
profiles are displayed for total thymocytes. Histograms show the expression of ZAP-YEEI and clonotypic TCR by cells from TCR-Tg ⫻ ZAPYEEI (black line) and TCR-Tg mice (u) during the transition from the DP
to the CD8 SP stage by gating in the CD4 vs CD8 dot plot. A, P14 TCR-Tg
mice. P14 TCR was detected with the anti-V␣2 Ab. B, Female HY TCR-Tg
mice. The clonotypic TCR is detected with the T3.70 Ab. C, Histogram
overlays compare the expression of HSA and CD5 on TCR-Tg ⫻ ZAPYEEI (black line) and TCR-Tg mice (u) in gated CD8 SP thymocytes.
7393
7394
TCR AVIDITY-DEPENDENT ADAPTATION OF CD8 T CELLS
Table I. Absolute numbers of CD8 T cells in TCR-Tg and dTg micea
Rag⫹/⫺ Backgroundb
Thymus
LN
Rag⫺/⫺ Backgroundc
Mice
Va2⫹CD8⫹
T3.70⫹CD8⫹
P14 CD8-SP
HY CD8-SP
ZAP-WT
ZAP-YEEI
ZAP-WT
ZAP-YEEI
ND
ND
9.18 ⫾ 1.22 (n ⫽ 3)
5.79 ⫾ 1.33 (n ⫽ 3)
ND
ND
5.43 ⫾ 2.37 (n ⫽ 4)
15.77 ⫾ 6.06 (n ⫽ 4)
17.62 ⫾ 9.14 (n ⫽ 9)
14.11 ⫾ 6.05 (n ⫽ 9)
8.44 ⫾ 3.05 (n ⫽ 4)
5.67 ⫾ 0.96 (n ⫽ 4)
10.14 ⫾ 2.53 (n ⫽ 5)
8.95 ⫾ 2.58 (n ⫽ 5)
1.08 (n ⫽ 4)d
2.66 (n ⫽ 3)d
a
Total thymocytes and LN cells from control (ZAP-WT) or dTg (ZAP-YEEI) mice were isolated and counted. The number of clonotypic TCRpositive, CD8-SP cells b or of CD8-SP cells c in each sample was calculated after assessing the percentage of these cells by FCM analysis.
d
These data were obtained by counting cells after pooling samples and dividing by indicated numbers of mice. Data represent mean ⫾ SE (number
⫻106).
Discussion
Proposed more than a decade ago on theoretical grounds, TCR
signal tuning is conceived as a cell-intrinsic device integrating selection-based mechanisms that maintain self-recognition and tolerance (7). However, when and how such tuning is achieved are
unclear. During thymocyte maturation, spMHC-generated signals
appear to trigger adjustments in expression levels of surface receptors, including CD5 and CD2, which, in turn, would regulate
TCR sensitivity (10, 11, 38). Although CD5 may play a similar
role in setting the activation threshold of peripheral naive T cells
(11–13), it is not known whether other receptors and/or signaling
proteins are involved in such a regulation. Moreover, modifications of T cell responsiveness resembling signal tuning have been
observed in CD4 T cells stimulated by chronic exposure to
Ag (16).
In this study we tested the signaling tuning capacity of naive T
cell challenged in vivo with increased excitation by self-stimuli.
FIGURE 7. Phenotypic characterization of peripheral CD8 T cells of
P14 and H-Y mice expressing ZAP-YEEI. FCM analysis of cells stained
with anti-clonotypic Abs conjugated to biotin-streptavidin-PerCP, antiCD8-allophycocyanin, and anti-CD5-PE and intracellularly stained with
anti-VSV-Alexa488. Histograms show the expression of ZAP-YEEI, clonotypic TCR and CD5 on TCR-Tg ⫻ ZAP-YEEI (black line) and TCR-Tg
mice (u) by gated CD8 T cells. The percentage of transgene-expressing
cells and the ratio of MFI of clonotypic TCR and CD5 of TCR-Tg vs that
of TCR-Tg ⫻ ZAP-YEEI mice are indicated.
This condition was achieved by targeting the expression of a gainof-function Zap70 mutant at the onset of CD8 SP thymocyte appearance and in mature CD8 T cells. Our data suggest that in mice
with a normal TCR repertoire, most naive CD8 T cells tune their
signaling machinery toward a higher activation threshold when
experiencing stronger and prolonged excitation after spMHC interaction, a scenario predicted by the tunable activation threshold
hypothesis (7). Consistent with signaling adaptation, ZAP-YEEIexpressing CD8 T cells showed no sign of activation in vivo, but,
on the contrary, displayed a lower TCR signaling status, as demonstrated by reduced in vitro cytokine secretion in response to
TCR/CD28 costimulation. Both TCR expression and responsiveness to stimulation were dynamically tuned at the single-cell level
and could be restored when cells were deprived of TCR/spMHC
interaction for several hours.
The apparently contradictory observation of a higher in vivo
excitation to spMHC, detected as basal LAT phosphorylation (Fig.
FIGURE 8. Proliferation of P14 CD8 T cells expressing ZAP-YEEI in
T lymphocyte-deficient hosts and expression IL7R ␣-chain in P14 ⫻ ZAPYEEI mice. A, Approximately 5 ⫻ 105 LN cells from P14Rag⫺/⫺ (䡺) and
P14Rag⫺/⫺ ⫻ ZAP-YEEI (f) mice were cotransferred into B6.CD3⑀⫺/⫺
hosts. The distinction between the two mouse populations was conducted
through intracellular staining with the anti-VSV Ab. Each point in the
graph represents the total number of CD8 cells recovered from the spleens
of the hosts (mean ⫾ SD of three mice per time point). B, FCM analysis
of cells stained with biotinylated anti-Va2 Ab-streptavidin-PerCP, antiCD8-allophycocyanin, and anti-IL7Ra-PE. Histograms show IL7R␣ expression on gated CD8 Va2⫹ T cells from P14 ⫻ ZAP-YEEI (black line)
and P14 mice (u). The ratio of MFI in P14 vs P14 ⫻ ZAP-YEEI mice is
indicated.
Downloaded from http://www.jimmunol.org/ by guest on June 15, 2017
fraction of cytokine-producing CD8 T cells in HY ⫻ ZAP-YEEI
mice was increased compared with control HY CD8 cells (Fig.
9B). Thus, despite the decrease in TCR levels and the increase in
the negative regulator CD5 on HY ⫻ ZAP-YEEI CD8 T cells,
their responsiveness to Ag stimulation is improved by ZAP-YEEI
expression.
The Journal of Immunology
4), and a lower in vitro responsiveness may be explained in several
ways. Cells on the brink of completing their TCR signal tuning
(e.g., recent thymic emigrants) and/or cells bearing TCRs with low
avidity for spMHC and improving their fitness by exploiting ZAPYEEI may both reveal a stronger spMHC-driven basal signal and
coexist with a majority of ZAP-YEEI-expressing cells having already tuned down their TCR signaling (lower responders). An alternative explanation is offered by the observation that ZAP-YEEIexpressing cells showed faster LAT and ERK-2 dephosphorylation
after TCR triggering (Fig. 4, B and C). Thus, augmented early
phosphorylation might occur in all ZAP-YEEI-expressing cells,
but full activation could not take place because of fast signal
dampening due to reduced TCR availability and/or increase in
other negative feedback mechanisms (see below).
Studies of P14 and HY TCR-Tg mice expressing ZAP-YEEI
supported the view that TCR signal tuning is largely dictated by
TCR avidity for self-ligands, Thus, although expression levels of
ZAP-YEEI protein in SP CD8 thymocytes of HY ⫻ ZAP-YEEI
and P14 ⫻ ZAP-YEEI were comparable (compare Fig. 6, A and
B), only the latter showed decreased TCR expression, correlating
with TCR avidity for spMHC. Moreover, HY ⫻ ZAP-YEEI developing thymocytes displayed accelerated expression of TCRhigh
cells (Fig. 6B), and their mature counterpart accumulates in the
periphery more than HY cells, just the opposite of the behavior of
P14 ⫻ ZAP-YEEI thymocytes and mature CD8 T cells.
The contrasting behavior of mature T cells induced by a similar
modification of their TCR signaling machinery is striking, but suggests that, depending on the initial strength of the TCR signal, the
increased reactivity induced by ZAP-YEEI may be over- or undercompensated. Thus, the strong signal generated by ZAP-YEEI
downstream of a high avidity or promiscuous TCR, such as P14,
would induce a negative feedback in thymocytes and/or peripheral
T cells, dampening their proliferation potential and limiting possible activation by self-ligand(s). On the contrary, the weak signal
generated by a low avidity TCR such as HY, although reinforced
by ZAP-YEEI, would still be compatible with positive selection
and normal survival/expansion in the periphery. Hence, although
some limited adaptation is induced through TCR and CD5 modulation, cells may acquire improved functional capacities.
Surprisingly, not all activation responses appeared to be equally
affected by signal tuning in ZAP-YEEI-expressing T cells. For
example, we never detected alterations in activation-induced upregulation of CD69 or CD25 on polyclonal or Tg TCR backgrounds (data not shown). Moreover, proliferation did not seem to
be affected in either SZ1 or P14 ⫻ ZAP-YEEI mice, whereas
HY ⫻ ZAP-YEEI CD8 T cells proliferated slightly faster than HY
T cells, as assessed by CSFE labeling (not shown). These unpredicted differences may be due to different activation thresholds of
these responses compared with cytokine production or to the different sensitivities of the in vitro assays used.
The unanticipated expression in TCR-Tg mice of ZAP-YEEI in
DP thymocyte provided a model for monitoring the fate of immature thymocytes subjected to more intense spMHC-induced signaling. Indeed, because Zap70 plays a pivotal role in both positive
and negative thymic selection (39), ZAP-YEEI expression in DP
may affect either event, or both, depending on TCR avidity for the
selecting ligand(s). Because we did not detect significant differences in thymic cellularity, subpopulation composition, or HSA
down-regulation, we argue that no gross alteration of positive and
negative selection was taking place. However, P14 and HY TCRbearing thymocytes were differently affected by ZAP-YEEI expression. On the one hand, TCRlow DP cells in P14 ⫻ ZAP-YEEI
mice were slightly increased, and SP TCRhigh cells were strongly
decreased, suggesting that modulation of TCR levels may avoid
negative selection when receiving strong spMHC stimuli. Similarly, a dramatic reduction of TCR levels in DP and TCRhigh SP
thymocytes was observed in P14 ⫻ CD5-deficient mice (11).
Moreover, TCRlow mature T cells appeared in the periphery of
these animals (11). However, DP and SP cells were decreased in
these mice, indicating that the absence of CD5 leads to higher TCR
signaling to spMHC than the expression of ZAP-YEEI. Hence, the
latter situation set the thymocytes in a state compatible with a
decrease in TCR expression rather than negative selection. In contrast, ZAP-YEEI expression in HY DP cells induced an earlier
appearance of TCRhigh cells at the DP and intermediate DP to SP
stages. However, this effect did not increase mature thymocyte
numbers compared with HY mice (compare HY and HY ⫻ ZAPYEEI mice in Table I). Taken together, our data reveal that signaling plasticity based on TCR level modulation operates concomitantly with selection processes. Accordingly, it has been recently
reported that surface levels of HY, 2C, and OT1 TCR-Tg cells
inversely correlate with increasing avidity for MHC class I (40).
Our observations suggest that TCR level modifications in response to augmented signaling may take place during all T cell
developmental stages, because a significant TCR reduction was
evident in peripheral T cells. Indeed, TCR down-modulation in
HY ⫻ ZAP-YEEI mice was detected only in mature T cells. Moreover, augmented TCR down-regulation was apparent in P14 ⫻
ZAP-YEEI T cells compared with SP thymocytes (from 2–3.5
times reduction compared with P14) despite similar ZAP-YEEI
protein expression levels. Although the mechanism leading to TCR
down-modulation in ZAP-YEEI-expressing cells is still unclear,
we found that the expression of at least one of the TCR complex
subunits, namely the TCR ␨-chain, was decreased only at the protein, not the mRNA, level (unpublished observations), indicating a
post-translational regulatory mechanism possibly involving protein degradation.
If stronger signaling upon spMHC contact brings about TCR
down-modulation in CD8 T cells, it can be predicted that reducing
Downloaded from http://www.jimmunol.org/ by guest on June 15, 2017
FIGURE 9. Ag-induced cytokine production by P14 ⫻ ZAP-YEEI and
HY-ZAP-YEEI CD8 T cells. Splenocytes from P14 (A) and HY (B) mice
expressing ZAP-YEEI were stimulated in culture for 20 h with the
gp33– 41 P14 TCR-specific peptide or the HY TCR-specific peptide (5 ⫻
10⫺8 M), respectively. Cells (1 ⫻ 106) were then stained with anti-CD8allophycocyanin and anti-clonotypic-TCR conjugated to biotin-streptavidin-PerCP, fixed, permeabilized, and stained for anti-cytokine-PE Abs.
Histograms show the production of IFN-␥, IL-2, and TNF-␣ of TCR-Tg
(䡺) or TCR-Tg ⫻ ZAP-YEEI (f) by gated CD8⫹/clonotypic TCR⫹ cells.
A representative experiment of four is shown.
7395
7396
Acknowledgments
We thank Dr. L. Samelson for providing the anti-LAT Ab; Dr. Hakan
Wennbo, for the generation of one ZAP-YEEI transgenic founder; Dr.
Nicolas Legrand for providing H2-Db knockout mice; Dr. Paulo Vieira for
the IL-7-containing supernatant; M.-C. Wagner for purifying the antiVSV-G mAb; Beatrice Corre for her precious technical help; Drs. Sylvie
Garcia, Antonio Bandeira, and Gordon Langsley for helpful discussions
and suggestions; and Wendy Houssin for her invaluable secretarial
assistance.
Disclosures
The authors have no financial conflict of interest.
References
1. Anderson, M. S., E. S. Venanzi, L. Klein, Z. Chen, S. P. Berzins, S. J. Turley,
H. von Boehmer, R. Bronson, A. Dierich, C. Benoist, et al. 2002. Projection of
an immunological self shadow within the thymus by the aire protein. Science 298:
1395–1401.
2. Anderton, S. M., and D. C. Wraith. 2002. Selection and fine-tuning of the autoimmune T-cell repertoire. Nat. Rev. Immunol. 2: 487– 498.
3. Bouneaud, C., P. Kourilsky, and P. Bousso. 2000. Impact of negative selection on
the T cell repertoire reactive to a self-peptide: a large fraction of T cell clones
escapes clonal deletion. Immunity 13: 829 – 840.
4. Walker, L. S., and A. K. Abbas. 2002. The enemy within: keeping self-reactive
T cells at bay in the periphery. Nat. Rev. Immunol. 2: 11–19.
5. Grossman, Z., and W. E. Paul. 1992. Adaptive cellular interactions in the immune
system: the tunable activation threshold and the significance of subthreshold responses. Proc. Natl. Acad. Sci. USA 89: 10365–10369.
6. Grossman, Z., and W. E. Paul. 2000. Self-tolerance: context dependent tuning of
T cell antigen recognition. Semin. Immunol. 12: 197–203.
7. Grossman, Z., and W. E. Paul. 2001. Autoreactivity, dynamic tuning and selectivity. Curr. Opin. Immunol. 13: 687– 698.
8. Grossman, Z., and A. Singer. 1996. Tuning of activation thresholds explains
flexibility in the selection and development of T cells in the thymus. Proc. Natl.
Acad. Sci. USA 93: 14747–14752.
9. Tarakhovsky, A., S. B. Kanner, J. Hombach, J. A. Ledbetter, W. Muller,
N. Killeen, and K. Rajewsky. 1995. A role for CD5 in TCR-mediated signal
transduction and thymocyte selection. Science 269: 535–537.
10. Azzam, H. S., A. Grinberg, K. Lui, H. Shen, E. W. Shores, and P. E. Love. 1998.
CD5 expression is developmentally regulated by T cell receptor (TCR) signals
and TCR avidity. J. Exp. Med. 188: 2301–2311.
11. Azzam, H. S., J. B. DeJarnette, K. Huang, R. Emmons, C. S. Park,
C. L. Sommers, D. El-Khoury, E. W. Shores, and P. E. Love. 2001. Fine tuning
of TCR signaling by CD5. J. Immunol. 166: 5464 –5472.
12. Kassiotis, G., R. Zamoyska, and B. Stockinger. 2003. Involvement of avidity for
major histocompatibility complex in homeostasis of naive and memory T cells.
J. Exp. Med. 197: 1007–1016.
13. Smith, K., B. Seddon, M. A. Purbhoo, R. Zamoyska, A. G. Fisher, and
M. Merkenschlager. 2001. Sensory adaptation in naive peripheral CD4 T cells.
J. Exp. Med. 194: 1253–1261.
14. Stefanova, I., J. R. Dorfman, and R. N. Germain. 2002. Self-recognition promotes
the foreign antigen sensitivity of naive T lymphocytes. Nature 420: 429 – 434.
15. Bhandoola, A., X. Tai, M. Eckhaus, H. Auchincloss, K. Mason, S. A. Rubin,
K. M. Carbone, Z. Grossman, A. S. Rosenberg, and A. Singer. 2002. Peripheral
expression of self-MHC-II influences the reactivity and self-tolerance of mature
CD4⫹ T cells: evidence from a lymphopenic T cell model. Immunity 17:
425– 436.
16. Singh, N. J., and R. H. Schwartz. 2003. The strength of persistent antigenic
stimulation modulates adaptive tolerance in peripheral CD4⫹ T cells. J. Exp.
Med. 198: 1107–1117.
17. Pelosi, M., V. Di Bartolo, V. Mounier, D. Mège, J.-M. Pascussi, E. Dufour,
A. Blondel, and O. Acuto. 1999. Tyrosine 319 in the interdomain B of ZAP-70
is a binding site for the SH2 domain of Lck. J. Biol. Chem. 274: 14229 –14237.
18. Di Bartolo, V., D. Mège, V. Germain, M. Pelosi, E. Dufour, F. Michel,
G. Magistrelli, A. Isacchi, and O. Acuto. 1999. Tyrosine 319, a newly identified
phosphorylation site of ZAP-70, plays a critical role in T cell antigen receptor
signaling. J. Biol. Chem. 274: 6285– 6294.
19. Chan, A. C., M. Iwashima, C. W. Turck, and A. Weiss. 1992. ZAP-70: a 70 kd
protein-tyrosine kinase that associates with the TCR ␨ chain. Cell 71: 649 – 662.
20. Chan, A. C., M. Dalton, R. Johnson, G.-H. Kong, T. Wang, R. Thoma, and
T. Kurosaki. 1995. Activation of ZAP-70 kinase activity by phosphorylation of
tyrosine 493 is required for lymphocyte antigen receptor function. EMBO J. 14:
2499 –2508.
21. Zhang, W., J. Sloan-Lancaster, J. Kitchen, R. P. Trible, and L. Samelson. 1998.
LAT: the ZAP-70 tyrosine kinase substrate that links T cell receptor to cellular
activation. Cell 92: 83–92.
22. Williams, B. L., B. J. Irvin, S. L. Sutor, C. C. S. Chini, E. Yacyshyn,
J. Bubeck Warderburg, M. Dalton, A. C. Chan, and R. T. Abraham. 1999. Phosphorylation of Tyr319 in ZAP-70 is required for T-cell antigen receptor-dependent phospholipase C-␥1 and Ras activation. EMBO J. 18: 1832–1844.
23. Gong, Q., X. Jin, A. M. Akk, N. Foger, M. White, G. Gong, J. B. Wardenburg,
and A. C. Chan. 2001. Requirement for tyrosine residues 315 and 319 within ␨
chain-associated protein 70 for T cell development. J. Exp. Med. 194: 507–518.
24. Songyang, Z., S. E. Shoelson, M. Chaudhuri, G. Gish, T. Pawson, W. G. Haser,
F. King, T. Roberts, S. Ratnofsky, R. J. Lechleider, et al. 1993. SH2 domains
recognize specific phosphopeptide sequences. Cell 72: 767–778.
25. Ellmeier, W., M. J. Sunshine, K. Losos, F. Hatam, and D. R. Littman. 1997. An
enhancer that directs lineage-specific expression of CD8 in positively selected
thymocytes and mature T cells. Immunity 7: 537–547.
26. Ellmeier, W., M. J. Sunshine, K. Losos, and D. R. Littman. 1998. Multiple developmental stage-specific enhancers regulate CD8 expression in developing thymocytes and in thymus-independent T cells. Immunity 9: 485– 496.
27. Kreis, T. E. 1986. Microinjected antibodies against the cytoplasmique domain of
vesicular stomatitis virus glycoprotein block its transport to the cell surface.
EMBO J. 5: 931–941.
28. Legrand, N., and A. A. Freitas. 2001. CD8⫹ T lymphocytes in double ␣␤ TCR
transgenic mice. I. TCR expression and thymus selection in the absence or in the
presence of self-antigen. J. Immunol. 167: 6150 – 6157.
29. Vosshenrich, C. A., A. Cumano, W. Muller, J. P. Di Santo, and P. Vieira. 2004.
Pre-B cell receptor expression is necessary for thymic stromal lymphopoietin
Downloaded from http://www.jimmunol.org/ by guest on June 15, 2017
or eliminating spMHC-induced signals would result in the recovery of TCR levels. We show in this study that this was actually the
case in ZAP-YEEI⫹ cells from SZ1 mice (Fig. 5), and that TCRmediated responses were also restored under these conditions. A
partial recovery of TCR expression was also observed in vivo upon
transfer of P14Rag⫺/⫺ and P14Rag⫺/⫺ ⫻ ZAP-YEEI mice in
CD3⑀⫺/⫺Db⫺/⫺ hosts. The initial 3.5-fold increased ratio in V␣2
TCR MFI between control and dTg cells was decreased to 2-fold
48 h after transfer (not shown). The incomplete recovery in this
setting may be due to several reasons: for instance, the presence of
MHC molecules on LN cells of donor origin or the expression of
other MHC haplotypes in Db⫺/⫺ hosts may limit TCR recovery by
P14 ⫻ ZAP-YEEI cells. Taken together, our results demonstrate
that TCR expression and T cell responsiveness may be dynamically regulated in peripheral T cells according to the strength of
continuous spMHC stimulation.
It is worth noting that despite similar or only slightly different
TCR levels in CD8 T cells from C57BL/6, P14, and HY T cells
expressing ZAP-YEEI (2-, 3.5-, and 1.7-fold, respectively), responsiveness was decreased in the former two, but enhanced in the
latter, suggesting that the TCR level is only one element contributing to hyporesponsiveness. Even though we attempted to identify
modifications of known positive or negative signaling regulators,
we found no changes in the expression levels of LAT, Lck, Fyn,
Src homology region 2 domain-containing phosphatase-1, and cCbl (Fig. 4C and data not shown) in cells expressing ZAP-YEEI.
Nonetheless, the faster dephosphorylation of LAT and ERK-2 occurring in these cells after TCR cross-linking suggests increased
recruitment/activation of negative regulators (e.g., the protein tyrosine phosphatase Src homology region 2 domain-containing
phosphatase-1) (41).
Another hallmark of TCR signal tuning is the modulation of
CD5 levels (11, 13). We observed that CD5 levels were reduced in
T cells manifesting lower responses in vitro and in vivo. In contrast, CD5 increased in T cells with improved responsiveness (e.g.,
HY ⫻ ZAP-YEEI T cells). Based on the negative regulation exerted by CD5 on TCR signaling (9), one would expect that an
increase in CD5 expression would decrease T cell reactivity and
vice versa. However, several observations have previously suggested that CD5 levels actually mirror the intensity of TCR-dependent signals (11, 13). Therefore, our results, rather, suggest that
in response to strong signal, T cells or thymocytes may first respond, if contextually necessary, by reducing TCR-directed signaling and then by modifying CD5 expression.
In conclusion, our novel developmental stage-specific system
for TCR signaling modification provides experimental evidence of
the existence of tuning mechanisms that allow adjustments in T
cell responsiveness to environmental cues and that would limit
potential autoreactivity.
TCR AVIDITY-DEPENDENT ADAPTATION OF CD8 T CELLS
The Journal of Immunology
30.
31.
32.
33.
34.
35.
responsiveness in the bone marrow but not in the liver environment. Proc. Natl.
Acad. Sci. USA 101: 11070 –11075.
van Meerwijk, J. P. M., and R. N. Germain. 1993. Development of mature CD8⫹
thymocytes: selection rather than instruction? Science 261: 911–915.
Liu, H., M. Rhodes, D. L. Wiest, and D. A. Vignali. 2000. On the dynamics of
TCR:CD3 complex cell surface expression and downmodulation. Immunity 13:
665– 675.
Huppa, J. B., M. Gleimer, C. Sumen, and M. M. Davis. 2003. Continuous T cell
receptor signaling required for synapse maintenance and full effector potential.
Nat. Immunol. 4: 749 –755.
Marrack, P., and J. Kappler. 2004. Control of T cell viability. Annu. Rev. Immunol. 22: 765–787.
Kisielow, P., H. Bluthmann, U. D. Staerz, M. Steinmetz, and H. von Boehmer.
1988. Tolerance in T-cell-receptor transgenic mice involves deletion of nonmature CD4⫹8⫹ thymocytes. Nature 333: 742–746.
Pircher, H., K. Burki, R. Lang, H. Hengartner, and R. M. Zinkernagel. 1989.
Tolerance induction in double specific T-cell receptor transgenic mice varies with
antigen. Nature 342: 559 –561.
7397
36. Legrand, N., and A. A. Freitas. 2001. CD8⫹ T lymphocytes in double ␣␤ TCR
transgenic mice. II. Competitive fitness of dual ␣␤ TCR CD8⫹ T lymphocytes in
the peripheral pools. J. Immunol. 167: 6158 – 6164.
37. von Boehmer, H. 1990. Developmental biology of T cells in T cell receptor
transgenic mice. Annu. Rev. Immunol. 8: 531–556.
38. Wong, P., G. M. Barton, K. A. Forbush, and A. Y. Rudensky. 2001. Dynamic
tuning of T cell reactivity by self-peptide-major histocompatibility complex ligands. J. Exp. Med. 193: 1179 –1187.
39. Negishi, I., N. Motoyama, K. Nakayama, S. Senju, S. Hatakeyama, Q. Zhang,
A. C. Chan, and D. Y. Loh. 1995. Essential role for ZAP-70 in both positive and
negative selection of thymocytes. Nature 376: 435– 438.
40. Kieper, W. C., J. T. Burghardt, and C. D. Surh. 2004. A role for TCR affinity in
regulating naive T cell homeostasis. J. Immunol. 172: 4044.
41. Kosugi, A., J. Sakakura, K. Yasuka, M. Ogata, and T. Hamaoka. 2001. Involvement of SHP-1 tyrosine phosphase in TCR-mediated signaling pathways in lipid
rafts. Immunity 14: 669 – 680.
Downloaded from http://www.jimmunol.org/ by guest on June 15, 2017