ArticlePDF Available

Cutting Edge: Programmed Death-1 Up-Regulation Is Involved in the Attrition of Cytomegalovirus-Specific CD8+ T Cells in Acute Self-Limited Hepatitis B Virus Infection

Authors:

Abstract and Figures

Attrition of heterologous virus-specific CD8(+) T cells has been demonstrated in murine viral infection; however, little is known regarding this phenomenon in human viral infections. In this study, we observed that CMV-specific CD8(+) T cells displayed numerical decline and functional impairment in the early phase of acute infection, whereas programmed death-1 (PD-1) expression was significantly up-regulated by these CMV-specific CD8(+) T cells. This early PD-1 up-regulation was found to be closely associated with the increased apoptotic sensitivity of CMV-specific CD8(+) T cells. The in vitro addition of anti-PD-1 further enhanced the spontaneous apoptosis of CMV-specific CD8(+) T cells; however, blockade of the PD-1-mediated pathway with anti-PD-L1 significantly restored the CMV-specific CD8(+) T cell proliferation and IFN-gamma production. Thus, PD-1 plays a crucial role in the attrition of CMV-specific CD8(+) T cells in acute hepatitis B virus infection, which in turn, influences the preexisting homeostatic virus-specific CD8(+) T cell pool.
Content may be subject to copyright.
BRIEF REPORT | SEPTEMBER 15 2008
Cutting Edge: Programmed Death-1 Up-Regulation Is Involved in the
Attrition of Cytomegalovirus-Specic CD8
+
T Cells in Acute Self-Limited
Hepatitis B Virus Infection
1
Ji-Yuan Zhang; ... et. al
J Immunol (2008) 181 (6): 3741–3744.
https://doi.org/10.4049/jimmunol.181.6.3741
Related Content
Cutting Edge: Attrition of Plasmodium-Specic Memory CD8 T Cells Results in Decreased Protection That Is Rescued by
Booster Immunization
J Immunol (April,2011)
Cross-Reactive Antigen Is Required to Prevent Erosion of Established T Cell Memory and Tumor Immunity: A Heterologous
Bacterial Model of Attrition
J Immunol (August,2002)
IFN-Induced Attrition of CD8 T Cells in the Presence or Absence of Cognate Antigen during the Early Stages of Viral
Infections
J Immunol (April,2006)
Downloaded from http://journals.aai.org/jimmunol/article-pdf/181/6/3741/1266063/zim01808003741.pdf by guest on 01 December 2023
Cutting Edge
Cutting Edge
Cutting Edge: Programmed Death-1 Up-Regulation Is
Involved in the Attrition of Cytomegalovirus-Specific
CD8
T Cells in Acute Self-Limited Hepatitis B
Virus Infection
1
Ji-Yuan Zhang,
2
Zheng Zhang,
2
Bo Jin, Shu-Ye Zhang, Chun-Bao Zhou,
Jun-Liang Fu, and Fu-Sheng Wang
3
Attrition of heterologous virus-specific CD8
T cells
has been demonstrated in murine viral infection; how-
ever, little is known regarding this phenomenon in hu-
man viral infections. In this study, we observed that
CMV-specific CD8
T cells displayed numerical de-
cline and functional impairment in the early phase of
acute infection, whereas programmed death-1 (PD-1)
expression was significantly up-regulated by these
CMV-specific CD8
T cells. This early PD-1 up-regu-
lation was found to be closely associated with the in-
creased apoptotic sensitivity of CMV-specific CD8
T
cells. The in vitro addition of anti-PD-1 further en-
hanced the spontaneous apoptosis of CMV-specific
CD8
T cells; however, blockade of the PD-1-mediated
pathway with anti-PD-L1 significantly restored the
CMV-specific CD8
T cell proliferation and IFN-
production. Thus, PD-1 plays a crucial role in the attri-
tion of CMV-specific CD8
T cells in acute hepatitis B
virus infection, which in turn, influences the preexisting
homeostatic virus-specific CD8
T cell pool. The
Journal of Immunology, 2008, 181: 3741–3744.
Memory T cells are capable of stable survival after the
resolution of a pathogenic infection, even in envi-
ronments lacking MHC Ags (1). However, the sta-
ble survival of memory CD8
T cells in mice can be disrupted
by subsequent heterologous viral or bacterial infections in
which the number of the majority of non-cross-reactive mem-
ory CD8
T cells decreases via cytokine-dependent mecha-
nisms (2–6). In mice, this phenomenon has been identified as
the attrition of memory CD8
T cells (7, 8). The following two
models have been proposed to explain this attrition: 1) the pas-
sive attrition model, which predicts that old memory T cells are
defeated by newly formed memory T cells while competing for
survival niches within the immune system; and 2) the active at-
trition model, which predicts that the preexisting memory cells
undergo direct apoptotic attrition (7–9). However, in humans
there is no report on the attrition of memory CD8
T cells, and
the mechanisms underlying such attrition remain to be
elucidated.
The interaction between programmed death-1 (PD-1)
4
and
its ligand PD-L1 has been reported to play a crucial role in in-
ducing T cell exhaustion (10, 11), anergy (12), and apoptosis
(13) in mice and human viral infections (14 –22). These studies
indicated that PD-1 up-regulation may exhaust virus-specific
CD8
T cells, facilitating viral persistence in chronic viral in-
fection. The in vitro blockade of PD-1/PD-L1 interaction re-
versed the exhaustion of the virus-specific T cells (10–12, 14
19). In acute viral infections, PD-1 was transiently up-regulated
on virus-specific CD8
T cells in the early phase of the infec-
tion, and successful clearance of these viruses often correlated
with decreased PD-1 expression and the development of func-
tional CD8
memory T cells (21, 22). Moreover, PD-1-posi-
tive virus-specific CD8
T cells showed higher sensitivity to
apoptosis in the case of HIV-1 infection (15), and higher in
vitro PD-1/PD-L1 interaction enhanced the apoptosis of hep-
atitis B virus (HBV)-specific CD8
T cells during acute HBV
(AHB) infection (21). These findings indicate that PD-1-me-
diated coinhibitory signaling not only inhibits the function but
also regulates the survival of virus-specific CD8
T cells during
acute and chronic viral infections. Thus far, little information is
available regarding the roles of PD-1 in the attrition of heter-
ogenous virus-specific CD8
T cells. Notably, a recent study
demonstrated that PD-1 is responsible for the depletion of au-
toreactive CD8
T cells in mice (13).
In this study, we investigated the association between PD-1
expression and the fate of CMV-specific CD8
T cells in acute
Research Center for Biological Therapy, Beijing 302 Hospital, Beijing, China
Received for publication April 28, 2008. Accepted for publication July 24, 2008.
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 study was supported by National Key Basic Research Program of China Grants
2007CB512805 and 2006CB504305 and National Science Fund for Distinguished
Young Scholars Grant 30525042.
2
J.-Y.Z. and Z.Z. contributed equally to this work.
3
Address correspondence and reprint requests to Dr. Fu-Sheng Wang, Research Center
for Biological Therapy, Beijing 302 Hospital, Beijing 100039, China. E-mail address:
fswang@public.bta.net.cn
4
Abbreviations used in this paper: PD-1, programmed death-1; AHB, acute hepatitis B;
HBV, hepatitis B virus; HC, healthy control; HBsAg, hepatitis B surface antigen; PD-L1,
PD-1 ligand.
Copyright © 2008 by The American Association of Immunologists, Inc. 0022-1767/08/$2.00
www.jimmunol.org
Downloaded from http://journals.aai.org/jimmunol/article-pdf/181/6/3741/1266063/zim01808003741.pdf by guest on 01 December 2023
self-limited HBV infection. HBV infection often results in
acute self-limited infection in adults with prominent clinical
presentation (20). The majority of the patients in this study had
previously been infected with CMV during early childhood and
established long-term immune protection against this infec-
tion. Our data indicate that PD-1 is involved in the attrition of
CMV-specific CD8
T cells in AHB, which, in turn, influ-
ences the preexisting homeostatic virus-specific CD8
T cell
pool.
Materials and Methods
Subjects
A total of 16 HLA-A2-positive AHB patients (12 male and four female; mean
age, 35.6 8.8 years) were diagnosed as described in our recent report (21). In
brief, the serum alanine aminotransferase levels at the clinical onset were at least
10-fold the upper limit of the normal level at the time of the first detection of
serum hepatitis B surface Ag (HBsAg) and Ig M anti-hepatitis B core Ag ac-
companying multispecific anti-HBV CD8
T cell responses (21). All of the
AHB patients finally recovered clinically and displayed alanine aminotransfer-
ase normalization and HBsAg seroconversion within 6 mo of the initial onset of
symptoms. Sixteen healthy subjects were recruited as controls (HCs). All par-
ticipants were anti-hepatitis A virus, anti-hepatitis C virus, anti-hepatitis D vi-
rus, and anti-HIV-1 and anti-HIV-2 Ab-negative. Informed consent was ob-
tained from each participant. The study was approved and has been reviewed by
the local medical ethics committee.
Abs and reagents
All Abs were purchased from BD Biosciences, except for the anti-
PD-L1-blocking mAb (MIH1) and the anti-human PD-1 Ab (AF 1086), which
were purchased from eBioscience and R&D Systems, respectively. A PE-labeled
CMV pentamer loaded with the pp65 epitope (495–503, NLVPMVATV) was
synthesized by ProImmune. Lymphocyte counts of these patients were deter-
mined using an automated differential blood count system. Plasma IFN-
con-
centration was measured using a standard ELISA kit from BioSource
International.
Apoptosis assay
Freshly isolated PBMCs were stained with the PE-labeled CMV pentamer, fol-
lowed by anti-CD8-PerCP and anti-PD-1-allophycocyanin and finally with
annexin V according to our previously reported protocols (21). In some exper-
iments, 1–1.5 10
6
PBMCs were cultured in 24-well plates with or without
plate-bound anti-human PD-1 Ab (20
g/ml) for 12 h at 37°C. The cells were
subsequently stained with annexin V to detect apoptosis.
Intracellular IFN-
staining assay
PBMCs were stimulated with pp65 495–504 peptide (10
g/ml) plus anti-
PD-L1 (10
g/ml) or isotype control Ab for 6 h. GolgiStop (BD PharMingen)
was added to the cells after stimulation for 2 h. Intracellular IFN-
staining
assay was performed according to our previously reported protocols (16, 21).
Ag-specific cell proliferation
CFSE-based proliferation assays were performed according to our previously
described protocols (16, 21). Cells were stimulated with pp65 495–504 peptide
(2
g/ml), anti-human CD28 (0.5
g/ml), and human rIL-2 (20 U/ml) in the
presence of anti-PD-L1 (10
g/ml) or isotype control Ab in a 24-well plate. On
day 6, the cells were supplemented with 0.2 ml of fresh medium containing the
above-mentioned cytokines. On day 10, the cells were stained with pentamers.
Statistical analysis
All data were analyzed using SPSS software. Comparison between various
groups was performed using the Mann-Whitney Utest. For the blockade assay,
statistical comparisons were performed using the Wilcoxon matched pairs ttest.
The correlation between variables was evaluated using the Spearman’s rank cor-
relation test. For all two-tailed tests, p0.05 was considered significant.
Results and Discussion
The attrition of preexisting virus-specific CD8
T cells follow-
ing heterologous viral infections has been well demonstrated in
mice models (2–9). However, whether this attrition occurs in
human viral infections is debatable; in particular, it is unknown
whether acute HBV infection affects the preexisting CMV-spe-
cific CD8
T cell pools in AHB patients. To address the issue,
we first found that both the percentages and absolute number of
CMV-specific CD8
T cells were significantly reduced at the
clinical onset of AHB in patients compared with those of HCs
(Fig. 1A). In contrast, multispecific anti-HBV CD8
T cell re-
sponses at such an early phase were observed in the patients
(21). Thus, these data primarily indicate that acute HBV infec-
tion may induce the attrition of CMV-specific CD8
T cells
during the early stage of HBV infection in humans. This early
attrition of CMV-specific CD8
T cells might make room for
newly formed HBV-specific T cells within the immune system
and facilitate the mounting of a strong anti-HBV T cell
response.
PD-1 has been demonstrated to regulate the survival of virus-
specific CD8
T cells during acute and chronic viral infections
(15, 21). To investigate the association between PD-1 expres-
sion and the attrition of CMV-specific CD8 T cells in AHB
infections, we serially analyzed the PD-1 expression on these
CMV-specific CD8 T cells. Our data indicated that PD-1 ex-
pression on CMV-specific CD8
T cells was significantly up-
regulated in AHB patients compared with that in HCs (Fig.
1B). The pool data further confirmed this observation (Fig.
1C). The underlying mechanism of PD-1 up-regulation on
these preexisting virus-specific CD8
T cells is not yet clear. A
study has suggested that PD-1 expression occurs before the ap-
pearance of early T cell activation markers, including CD69
and CD25, and that PD-1 expression may be controlled by
early responding cytokines in addition to TCR signaling (12).
In this regard, CMV was not observed to be reactivated, and no
cross-reactivity was observed between CMV and HBV in these
AHB patients. Interestingly, we found that PD-1 was also up-
regulated by influenza-specific CD8
T cells in AHB patients
FIGURE 1. Attrition of CMV-specific CD8
T cells at the clinical onset of
acute HBV infection. A, The percentages and numbers of CMV-pp65 CD8
T
cells in AHB patients and HCs. The numbers of circulating CMV-pp65 CD8
T cells were calculated as the lymphocyte count (cells/microliter) percentage
of CD8 percentage of CMV-pp65 pentamer. B, Representative dot plots of
PD-1 expression on CMV-pp65 CD8
T cells. Values in the upper right quad-
rants indicate the percentages of CMV-pp65 CD8
T cells that express PD-1.
C, Pool data of PD-1 expression on CMV-pp65 CD8
T cells. D, Pool data of
the plasma IFN-
concentration.
3742 CUTTING EDGE: PD-1-MEDIATED MEMORY CD8 T CELL ATTRITION
Downloaded from http://journals.aai.org/jimmunol/article-pdf/181/6/3741/1266063/zim01808003741.pdf by guest on 01 December 2023
compared with those of HCs (data not shown). Thus, PD-1
up-regulation on CMV-specific CD8 T cells seems to be less
likely due to CMV-specific TCR signaling, provided that influ-
enza infection does not result in a viral persistence. Therefore,
further studies on the factors inducing PD-1 up-regulation on
CMV-specific CD8 T cells are warranted.
Notably, there is a difference in the time at which the re-
sponses were observed in the AHB patients of this study and in
the study performed on mice models. The IFN-induced attri-
tion observed in mice models occurred at the first few days of
infection before Ag-specific T cells in response to the acute in-
fection; this attrition can be easily detected (2–9). By contrast,
AHB patients are usually identified between 10 and 15 wk after
HBV infection, when they become clinically symptomatic;
HBV-specific T cells are detectable at this stage (21). Thus, al-
though we also found that the serum IFN-
level was signifi-
cantly higher at clinical onset of infection than that in healthy
subjects (Fig. 1D), this time difference suggested that the attri-
tion of CMV-specific CD8 T cells in AHB patients may be a
different phenomenon than the IFN-induced attrition ob-
served in mice. Future studies should elucidate the mechanisms
responsible for the attrition in AHB patients.
PD-1 was originally cloned from apoptotic cell lines (22)
and could increase the apoptotic sensitivity of HBV-specific
CD8 T cells in acute HBV infections (21). Subsequently, we
examined whether PD-1 up-regulation affects the numbers
of CMV-specific CD8
T cells in AHB patients. Both PD-
1
high
and PD-1
median
CMV-specific CD8
T cell subsets
were observed to possess higher levels of annexin V staining
than the PD-1
low
subsets (Fig. 2A), suggesting that the
higher levels of PD-1 expression were associated with the
greater apoptosis sensitivity of CMV-specific CD8
T cells.
More importantly, we observed that plate-bound stimula-
tory anti-PD-1 directly enhanced the apoptosis of CMV-
specific CD8
T cells. Pooled data confirmed that the re-
sults were consistent for seven of nine patients (Fig. 2B). We
also longitudinally analyzed the association between PD-1
expression and the depletion of CMV-specific CD8
T cells
in seven AHB patients and found that the decreases in CMV-
specific CD8
T cells was positively correlated with the re-
duction in PD-1-expressing CMV-specific CD8
T cells
(Fig. 2C). These results indicate that PD-1 up-regulation
might mediate the apoptosis of CMV-specific CD8
T cells
in acute HBV infections.
FIGURE 2. PD-1 up-regulation is associated with the apoptosis of CMV-specific CD8
T cells in AHB patients. A, Representative histograms depict the annex-
in V positivity in PD-1
high
, PD-1
median
, and PD-1
low
CMV-pp65 CD8
T cells at the clinical onset in an AHB patient. Pool data indicate the percentages of annexin
V-positive cells in different subsets of CMV-pp65 CD8
T cell populations of 10 AHB patients. Horizontal bars present the median percentages of annexin
V-positive cells. B, Representative dot plots of annexin V staining in CMV-pp65 CD8
T cells in the presence and absence of plate-bound anti-PD-1 Ab (aPD-1).
Values in the upper right quadrants indicate the percentages of annexin V-positive CMV-pp65 CD8
T cells. The fold change in the percentage of CMV-pp65 CD8
T cells is shown. C, Representative dot plots represent the kinetics of PD-1 expression on CMV-pp65 CD8
T cells in acute HBV infections. The correlation
between the decrease in CMV-pp65 CD8
T cells and the reduction in PD-1-positive CMV-pp65 CD8
T cells was analyzed. Pen, Pentamer.
3743The Journal of Immunology
Downloaded from http://journals.aai.org/jimmunol/article-pdf/181/6/3741/1266063/zim01808003741.pdf by guest on 01 December 2023
Persistent PD-1 up-regulation can exhaust virus-specific T
cells during established chronic HIV, hepatitis C virus, and
HBV infections (10–19). In this section, we further
investigated whether the interference of PD-1 with PD-L1 in-
fluenced the function of CMV-specific CD8
T cells following
stimulation with a cognate peptide. We found that blockade of
PD-1/PD-L1 interaction by using an anti-PD-L1 blocking Ab
significantly increased IFN-
production (Fig. 3, Aand C) and
the accumulation of CFSE
low
pentamers (Fig. 3, Band D)
among these CMV-specific CD8
T cells. However, due to the
high apoptotic sensitivity of the PD-1
high
CMV-specific CD8
T cells, this expansion of CMV-specific CD8
T cells in the
presence of anti-PD-L1 may be a direct result of enhanced
CD8
T cell proliferation, decreased apoptosis, or a combina-
tion of the two effects. These data indicated that PD-1 may also
participate in the functional attrition of CMV-specific CD8
T
cells in acute HBV infections.
In summary, this study provides the first indication that
PD-1 up-regulation, at least in part, contributes to the attrition
of CMV-specific CD8
T cells in acute HBV infections. These
findings, therefore, extended the attrition of memory T cells
from mouse viral infections to human viral infections and pre-
sented a mechanism that influences the homeostasis in the pre-
existing virus-specific CD8
T cell pool.
Acknowledgments
All authors express sincere thanks to all HBV-infected individuals and healthy
participants in this study.
Disclosures
The authors have no financial conflict of interest.
References
1. Murali-Krishna, K., L. L. Lau, S. Sambhara, F. Lemonnier, J. Altman, and R. Ahmed.
1999. Persistence of memory CD8 T cells in MHC class I-deficient mice. Science 286:
1377–1381.
2. Selin, L. K., M. Y. Lin, K. A. Kraemer, D. M. Pardoll, J. P. Schneck, S. M. Varga,
P. A. Santolucito, A. K. Pinto, and R. M. Welsh. 1999. Attrition of T cell memory:
selective loss of LCMV epitope-specific memory CD8 T cells following infections
with heterologous viruses. Immunity. 11: 733–742.
3. McNally, J. M., C. C. Zarozinski, M. Y. Lin, M. A. Brehm, H. D. Chen, and
R. M. Welsh. 2001. Attrition of bystander CD8 T cells during virus-induced T-cell
and interferon responses. J. Virol. 75: 5965–5976.
4. Kim, S. K., and R. M. Welsh. 2004. Comprehensive early and lasting loss of memory
CD8 T cells and functional memory during acute and persistent viral infections. J. Im-
munol. 172: 3139–3150.
5. Smith, D. K., R. Dudani, J. A. Pedras-Vasconcelos, Y. Chapdelaine, H. van Faassen,
and S. Sad. 2002. Cross-reactive antigen is required to prevent erosion of established
T cell memory and tumor immunity: a heterologous bacterial model of attrition. J. Im-
munol. 169: 1197–1206.
6. Bahl, K., S. K. Kim, C. Calcagno, D. Ghersi, R. Puzone, F. Celada, L. K. Selin, and
R. M. Welsh. 2006. Interferon-induced attrition of CD8 T cells in the presence or
absence of cognate antigen during the early stages of viral infections. J. Immunol. 176:
4284–4295.
7. Selin, L. K., M. Cornberg, M. A. Brehm, S. Kim, C. Calcagno, D. Ghersi, R. Puzone,
F. Celada, and R. M. Welsh. 2004. CD8 memory T cells: cross-reactivity and heter-
ologous immunity. Semin. Immunol. 16: 335–347.
8. Selin, L. K., M. A. Brehm, Y. A. Naumov, M. Cornberg, S. Kim, S. C. Clute, and
R. M. Welsh. 2006. Memory of mice and men: CD8
T-cell cross-reactivity and
heterologous immunity. Immunol. Rev. 211: 164–181.
9. Jiang, J., L. L. Lau, and H. Shen. 2003. Selective depletion of nonspecific T cells
during the early stage of immune responses to infection. J. Immunol. 171: 4352– 4358.
10. Barber, D. L., E. J. Wherry, D. Masopust, B. Zhu, J. P. Allison, A. H. Sharpe,
G. H. Freeman, and R. Ahmed. 2006. Restoring function in exhausted CD8 T cells
during chronic viral infection. Nature 439: 682–687.
11. Sang-Jun, H., S.N. Mueller, E. J. Wherry, D. L. Barber, R. D. Aubert, A. H. Sharpe,
G. J. Freeman, and R. Ahmed. 2008. Enhancing therapeutic vaccination by blocking
PD-1-mediated inhibitory signals during chronic infection. J. Exp. Med. 205:
543–555.
12. Tsushima, F., S. Yao, T. Shin, A. Flies, S. Flies, H. Xu, K. Tamada, D. M. Pardoll, and
L. Chen. 2007. Interaction between B7–H1 and PD-1 determines initiation and re-
versal of T-cell anergy. Blood 110: 180–185.
13. Lin, S. J., C. D. Peacock, K. Bahl, and R. M. Welsh. 2007. Programmed death-1
(PD-1) defines a transient and dysfunctional oligoclonal T cell population in acute
homeostatic proliferation. J. Exp. Med. 204: 2321–2333.
14. Day, C. L., D. E. Kaufmann, P. Kiepiela, J. A. Brown, E. S. Moodley, S. Reddy,
E. W. Mackey, J. D. Miller, A. J. Leslie, C. DePierres, et al. 2006. PD-1 expression on
HIV-specific T cells is associated with T-cell exhaustion and disease progression. Na-
ture 443: 350–354.
15. Petrovas, C., J. P. Casazza, J. M. Brenchley, D. A. Price, E. Gostick, W. C. Adams,
M. L. Precopio, T. Schacker, M. Roederer, et al. 2006. PD-1 is a regulator of virus
specific CD8
T cell survival in HIV infection. J. Exp. Med. 203: 2281–2292.
16. Zhang, J. Y., Z. Zhang, X. Wang, J. L. Fu, J. Yao, Y. Jiao, L. Chen, H. Zhang, J. Wei,
L. Jin, et al. 2007. PD-1 up-regulation is correlated with HIV-specific memory CD8
T-cell exhaustion in typical progressors but not in long-term nonprogressors. Blood
109: 4671–4678.
17. Penna, A., M. Pilli, A. Zerbini, A. Orlandini, S. Mezzadri, L. Sacchelli, G. Missale,
and C. Ferrari. 2007. Dysfunction and functional restoration of HCV-specific CD8
responses in chronic hepatitis C virus infection. Hepatology 45: 588–601.
18. Boni, C., P. Fisicaro, C. Valdatta, B. Amadei, P. Di Vincenzo, T. Giuberti,
D. Laccabue, A. Zerbini, A. Cavalli, G. Missale, et al. 2007. Characterization of hep-
atitis B virus (HBV)-specific T-cell dysfunction in chronic HBV infection. J. Virol. 81:
4215–4225.
19. Chen, L., Z. Zhang, W. Chen, Z. Zhang, Y. Li, M. Shi, J. Zhang, L.Chen, S. Wang,
and F. S. Wang. 2007. B7–H1 up-regulation on myeloid dendritic cells significantly
suppresses T cell immune function in patients with chronic hepatitis B. J. Immunol.
178: 6634–6641.
20. Rehermann, B., and M. Nascimbeni. 2005. Immunology of hepatitis B virus and hep-
atitis C virus infection. Nat. Rev. Immunol. 5: 215–229.
21. Zhang, Z., J. Y. Zhang, E. J. Wherry, B. Jin, B. Xu, Z. S. Zou, S. Y. Zhang, B. S. Li,
H. F. Wang, H. Wu, et al. 2008. Dynamic programmed death-1 expression on virus-
specific CD8 T cells correlates with the outcome of acute hepatitis B. Gastroenterology
134: 1938–1949.
22. Ishida, Y., Y. Agata, K. Shibahara, and T. Honjo. 1992. Induced expression of PD-1,
a novel member of the immunoglobulin gene super family, upon programmed cell
death. EMBO J. 11: 3887–3895.
FIGURE 3. PD-1 up-regulation impairs the function of CMV-specific
CD8
T cells in the early phase of acute HBV infection. A, Representative dot
plots represent the intracellular IFN-
staining of CMV-pp65 CD8
T cells in
an AHB patient at the clinical onset. Values in the upper right quadrants indicate
the percentages of IFN-
-producing CD8
T cells. B, Representative dot plots
represent CFSE staining in an AHB patient. Values in the upper left quadrant
indicate the percentage of CFSE
low
pentamers. (C) Summary data for the fold
change in the percentage of IFN-
-producing CMV-pp65 CD8
T cells are
shown. D, Summary data for the fold change in the percentage of CFSE
low
pentamers are shown. Horizontal bars in Cand Dindicate the median fold
changes. aPD-11, anti-PD-11; Pen, pentamer.
3744 CUTTING EDGE: PD-1-MEDIATED MEMORY CD8 T CELL ATTRITION
Downloaded from http://journals.aai.org/jimmunol/article-pdf/181/6/3741/1266063/zim01808003741.pdf by guest on 01 December 2023
... There are limited reports on IL-10 secretion by CD8 + T cells in HCMV infection but liver transplant recipients who progressed to CMV disease had increased expression of PD-1 on both CD4 + and CD8 + T cells and increased IL-10 in their plasma [41]. Similarly, HCMV-specific CD8 + T cells in HBV-infected patients show increased PD-1 expression and loss of IFNγ production [42]. ...
... There has been extensive characterization of the HCMV-specific CD8 + T cell response to HCMV infection but few studies have addressed whether the secretion of IL-10 also occurs. The few reports of HCMV-specific IL-10 CD8 + T cell or regulatory responses have been in patients with other infections such as HIV [61] and HBV [42], or in liver transplant patients who have progressed to CMV disease [41], or in ageing studies using a pp65-specific tetramer [62]. There are also examples in the mouse model of cytomegalovirus (MCMV) of IL-10 production by CD8 + T cells; they have been identified in the salivary gland and in liver-infiltrating lymphocytes using both cytokine staining and IL-10 reporter mice [63][64][65][66]. ...
... The major phenotypes identified in infectious diseases include CD28-negative CD39-positive CD8 + T cells in HIV infection [36], and the upregulation of the inhibitory receptor PD-1 [58] on IL-10-secreting cells has also been observed in HIV, HBV and HCV [38][39][40]. The expression of PD-1 in CMV-specific CD4 + T cells has also been shown to suppress the production of anti-viral cytokines [59], and CMV-specific CD8 + T cells in HBV patients had increased PD-1 which correlated with the loss of IFNγ [42]. The assessment of the phenotype of CMV-specific IL-10-secreting CD8 + T cells in our study revealed an interesting and somewhat unique phenotype of an effector memory population expressing CD45RA which had a CD28 + CD39 + phenotype-this contrasts with other reports described for HIV [36]. ...
Article
Full-text available
HCMV-specific CD8+ T-cells are potent anti-viral effector cells in HCMV infected individuals, but evidence from other viral infections suggests that CD8+ T-cells can also produce the immunomodulatory cytokine IL-10. In this work we show that there are HCMV-specific IL-10 CD8+ T-cell responses in a cohort of individuals aged 23–76 years of age, predominantly directed against the HCMV proteins known to be expressed during latent infections as well as towards the proteins US3 and pp71. The analysis of HCMV-specific responses established during primary infection has shown that the IL-10 responses to US3 and pp71 HCMV proteins are detectable in the first weeks post infection, but not the responses to latency-associated proteins, and this IL-10 response is produced by both CD8+ and CD4+ T-cells. Phenotyping studies of HCMV-specific IL-10+ CD8+ T-cells show that these are CD45RA+ effector memory cells and co-express CD28 and CD57, however, the expression of the inhibitory receptor PD-1 varied from 90% to 30% between donors. In this study we have described for the first time the HCMV-specific IL-10 CD8+ T-cell responses and have demonstrated their broad specificity and the potential immune modulatory role of the immune response to HCMV latent carriage and periodic reactivation.
... Progressive dysfunction of the T-cell response is known to be a key element in the course of chronic viral infections such as human immunodeficiency virus and hepatitis C virus. 25 Similarly, the persistence of CMV increases the number of dysfunctional CMV-specific T cells over time [26][27][28][29] that are continuously driven to exhaustion, 28,30 eventually causing their elimination by apoptosis. 30 Programmed cell death 1 (PD-1) has been revealed to be a crucial player in the dysfunction of human immunodeficiency virus-specific T cells [31][32][33] and possibly of human CMV-specific CD8 þ T cells. 30 Moreover, PD-1 has been found to be involved in the clinical control of CMV infections after KTx and LTx. ...
... 30 Programmed cell death 1 (PD-1) has been revealed to be a crucial player in the dysfunction of human immunodeficiency virus-specific T cells [31][32][33] and possibly of human CMV-specific CD8 þ T cells. 30 Moreover, PD-1 has been found to be involved in the clinical control of CMV infections after KTx and LTx. 34,35 An intronic single nucleotide polymorphism (SNP) termed "PD-1.3 ...
Article
Full-text available
Background: Cytomegalovirus (CMV) has a role in chronic rejection and graft loss in kidney transplant (KTx) and lung transplant (LTx) recipients. In addition, donor CMV seropositivity is an independent risk factor for renal graft loss. The anti-CMV response might modulate this risk. Expression of programmed cell death 1 (PD-1), a receptor involved in viral-specific T-cell exhaustion, is influenced by a single nucleotide polymorphism called PD-1.3 (wild-type allele G, variant allele A). Methods: We performed a retrospective study to assess the impact of PD-1.3 on graft outcome in donor CMV seropositive (D+) and donor CMV seronegative (D-) KTx and LTx. We also performed a case-control study to evaluate the anti-CMVpp65 response according to genotype. Results: PD-1.3 was determined in 1,119 KTx and 181 LTx. In 481 D+ KTx, A allele carriers (24%) experienced significantly less graft failure compared with GG carriers (p = 0.001). Multivariate analysis showed that this association was independent of donor and recipient age, acute rejection episodes, and number of human leukocyte antigen mismatches (hazard ratio, 0.381; 95% confidence interval, 0.209-0.696; p = 0.002). Analysis in 85 D+ LTx showed similar results: A allele carriers had better survival (hazard ratio, 0.302; 95% confidence interval, 0.128-0.716; p = 0.006) and greater 6-month forced expiratory volume (71% ± 17% vs 54% ± 16%, p = 0.001). In D- recipients, PD-1.3 did not affect KTx or LTx outcome. Finally, AA recipients had a stronger anti-CMVpp65 T-cell response than matched GG recipients (p = 0.003). Conclusions: The A variant allele in PD-1.3 single nucleotide polymorphism improved graft survival in kidney and lung transplant recipients receiving grafts from CMV-positive donors.
... Interactions between the coinfecting pathogens and the host may alter the pathogenesis and transmission of one or both pathogens [1], disease progression [2] and control of multiple diseases. Immunological effects of mixed infection include by stander immune activation in the absence of cognate antigens [3] and cellular attrition [4]. ...
Preprint
Full-text available
Fishing communities surrounding Lake Victoria in Uganda show an HIV-1 prevalence of 28% and incidence rates of 5%. More than 50% of the fishermen on the shores of Lake Victoria are infected with S. mansoni . Fishermen are more likely to die of AIDS related illness than farmers in the Lake Victoria region. Using polychromatic flow cytometry and mesoscale discovery platform, HIV specific and non-specific responses were measured and compared within individuals when HIV and S. mansoni coinfected and after the S. mansoni was cleared. Sixty-two unique clusters of cells in the UMAP space were identified after stimulations with GAG PTE POOL-1 and GAG PTE POOL-2 independently. However, the frequency of only three clusters is significantly higher after S. mansoni clearance. In addition, S. mansoni infection is associated with higher IL-9 and IL-10 and lower IL-15 in HIV and S. mansoni coinfected individuals. IL-9 concentration at enrolment visit predicts CD4 decline. S. mansoni infection negatively affects HIV specific and non-specific immune responses in HIV and S. mansoni coinfected individuals.
... Engagement of PD-1 with its ligands causes exhaustion in T cells, resulting in blunting of T cell responses. A number of studies with various pathogens have reported that PD-1/PD-L interactions result in the inhibition of T and B cell proliferation [26][27][28][29] . Conversely, abrogation of such receptor/ligand interactions restores T cell functions and confers resistance against various pathogens. ...
Article
Full-text available
Malaria remains a major public health problem worldwide. The immune mechanisms that mediate protection against malaria are still unclear. Previously, we reported that mesenchymal stem cells (MSCs) play a critical role in host protection against malaria by altering the dynamic balance of T regulatory cells and effector T cells producing inflammatory cytokines. Here, we report that MSCs reprogram haematopoiesis in primary (bone marrow) and secondary (spleen) lymphoid organs to provide host protection against malaria. Adoptive transfer of MSCs from malaria-infected mice to naïve recipient mice that were subsequently infected with malaria parasites dramatically accelerated the formation of colony-forming units-erythroid cells in the bone marrow. Adoptively transferred MSCs also induced expression of the key erythroid cell differentiation factor GATA-1 in the spleen of recipient animals. Interestingly, we further observed a subtle increase in the CD34 + hematopoietic stem and progenitor cells in lymphoid organs, including spleen and lymph nodes. Infusion of MSCs also enhanced T cell proliferation, resulting in increased numbers of both CD4 + and CD8 + T cells in the spleen. MSCs also inhibited the induction of the negative co-stimulatory receptor programmed death-1 by T cells in recipient animals upon infection with malaria parasites. Taken together, our findings suggest that MSCs play a critical role in host protection against malaria infection by modulating erythropoiesis and lymphopoiesis.
... Negative regulation of T-cells may be associated with the level of PD-1 secreted on their surface. 21,22 . Negative signaling via PD-1 is associated with regulation of homeostatic T-cell proliferation in lymphopenic conditions 23 and prevention of autoimmunity during peripheral tolerance. ...
Article
Full-text available
It is suggested that programmed cell death protein-1 (PD-1) is involved in hepatitis B virus (HBV) infection, the leading cause of hepatocellular carcinoma globally. This study was multi-aimed, that is, to investigate the role of microRNA (miR) 4717 and its target, PD-1 and to determine how the rs10204525 polymorphism in the 3′ untranslated region (3′UTR) of PD-1 affects its interaction with miR-4717. The expression levels of miR-4717 with various single-nucleotide polymorphisms were measured by reverse transcription–quantitative polymerase chain reaction (RT-qPCR). A total of 54 tissue samples from HBV-infected individuals were collected, genotyped, and categorized into three groups; AA (n = 32), AG (n = 18), and GG (n = 4). The expression levels of gene PDCD1 and its corresponding PD-1 protein were significantly declined in the AA group as compared to AG and GG groups. There was a negative linear association between PDCD1 and miR-4717 in the tissue samples. HEPG2 cells transfected with an miR-4717 mimic or PD-1 small interfering (si)RNA exhibited significantly reduced expression levels of PDCD1 and PD-1, whereas cells transfected with an inhibitor of miR-4717 demonstrated greater expression levels of PDCD1 and PD-1 compared with the scramble control. In addition, cell viability and apoptosis were assessed in cells transfected with an miR-4717 mimic, PD-1 siRNA, or an miR-4717 inhibitor. Results revealed that treatment with the miR-4717 mimic or PD-1 siRNA enhanced viability of cells and reduced apoptosis. The results of this study suggest that rs10204525 polymorphism interferes with the interaction between PD-1 and miR-4717 and therefore induces apoptosis in liver cancer cells.
... In a mouse model of heterologous CMV/HBV infection, PD1 suppression was found to be upregulated by CMV-specific CD8 ϩ T cells, which was associated with enhanced apoptotic activity of these cells. Blockade of the PD1 pathway by anti-PDL1 antibody restored the proliferation and cytokine secretion by these CMV-specific CD8 ϩ T cells (422). Thus, besides type 1 IFN, PD1-mediated apoptosis is yet another mechanism proposed for attrition. ...
Article
Full-text available
Coinfections involving viruses are being recognized to influence the disease pattern that occurs relative to that with single infection. Classically, we usually think of a clinical syndrome as the consequence of infection by a single virus that is isolated from clinical specimens. However, this biased laboratory approach omits detection of additional agents that could be contributing to the clinical outcome, including novel agents not usually considered pathogens. The presence of an additional agent may also interfere with the targeted isolation of a known virus. Viral interference, a phenomenon where one virus competitively suppresses replication of other coinfecting viruses, is the most common outcome of viral coinfections. In addition, coinfections can modulate virus virulence and cell death, thereby altering disease severity and epidemiology. Immunity to primary virus infection can also modulate immune responses to subsequent secondary infections. In this review, various virological mechanisms that determine viral persistence/exclusion during coinfections are discussed, and insights into the isolation/detection of multiple viruses are provided. We also discuss features of heterologous infections that impact the pattern of immune responsiveness that develops.
... A similar phenomenon has been reported in humans as well. Patients acutely infected with hepatitis B virus were found to have reduced number of cytomegalovirus-specific CD8 T cells than the healthy (hepatitis B negative) cohort (Zhang et al., 2008). ...
Article
The immune response to an infection is determined by a number of factors, which also affect the generation of memory T cells afterwards. The immune response can also affect the stability of the pre-existing memory populations. The memory developed after an infection can influence the response to subsequent infections with unrelated pathogens. This heterologous immunity may deviate the course of disease and alter the disease outcome. The generation and stability of memory CD8 T cells and the influence of the history of infections on subsequent heterologous infections are studied in this thesis using different viral infection sequences. Previous studies using mice lacking individual immunoproteasome catalytic subunits showed only modest alterations in the CD8 T cell response to lymphocytic choriomeningitis virus (LCMV). In this study, I found that the CD8 T cell response to LCMV was severely impaired in mice lacking all three catalytic subunits of the immunoproteasome, altering the immunodominance hierarchy of the CD8 T cell response and CD8 T cell memory. Adoptive transfer experiments suggested that both inefficient antigen presentation and altered T cell repertoire contribute to the reduction of the CD8 T cell response in the immunoproteasome knockout mice. Immune responses generated during infections can reduce pre-existing memory T cell populations. Memory CD8 T cells have been shown to be reduced by subsequent heterologous infections. In this study, I re-examined the phenomenon using immune mice infected with LCMV, murine cytomegalovirus (MCMV) and vaccinia virus (VACV) in different infection sequences. I confirmed that memory CD8 T cells were reduced by heterologous infections, and showed that LCMV-specific memory CD4 T cells were also reduced by heterologous infections. Reduction of the memory CD8 T cells is thought to be the result of apoptosis of memory CD8 T cells associated with the peak of type I interferon early during infection. I showed that memory CD4 T cells were similarly driven to apoptosis early during infection; however, Foxp3+ CD4+ regulatory T cells were relatively resistant to virus infection-induced apoptosis, and were stably maintained during LCMV infection. The stability of Treg cells during viral infections may explain the relatively low incidence of autoimmunity associated with infections. The history of infections can deviate the course of disease and affect the disease outcome, but this heterologous immunity is not necessarily reciprocal. Previous studies have shown the effects of heterologous immunity during acute infections. In this thesis, I showed that the history of LCMV infection led to higher viral titers during persistent MCMV infection, caused more severe immunopathology at the beginning of infection, and reduced the number of MCMV-specific inflationary memory CD8 T cells after the period of memory inflation. In a different context of infection, the history of LCMV infection can be beneficial. LCMV-immune mice have been shown to have lower viral titers after VACV infection, but VACV-immune mice are not protected during LCMV infection. I found that memory CD8 T cells generated from LCMV and VACV infections were phenotypically different, but the differences could not explain the nonreciprocity of heterologous immunoprotection. By increasing the number of crossreactive VACV A11R198-205-specific memory CD8 T cells, however, I showed that some VACV-immune mice displayed reduced viral titers upon LCMV challenge, suggesting that the low number of potentially cross-reactive CD8 T cells in VACV-immune mice may be part of the reasons for the non-reciprocity of immunoprotection between LCMV and VACV. Further analysis deduced that both number of potentially cross-reactive memory CD8 T cells and the private specificity of memory CD8 T cell repertoire played a part in determining the outcome of heterologous infections.
Article
Full-text available
Pregnant mice infected with Lymphocytic Choriomeningitis Virus (Armstrong) (LCMV-Arm) experience high viral loads in the placenta and uterine tissue by 5–8 days post-infection, a time when the virus is nearly completely cleared from the spleen and blood. Interleukin 10 (IL-10) plays a crucial role in T cell responses associated with systemic viral clearance. Using the LCMV-arm model of infection, we examined first, whether IL-10 is involved in viral clearance in the placenta and uterine tissue and secondly, the potential mechanisms underlying this phenomenon. C57BL/6 (WT) and mice deficient in IL-10 (IL-10 KO) were infected with LCMV-Arm on day 10 of pregnancy. Placenta and uterine tissue, collected 2 and 8 days later, were analyzed using real time RT-PCR, plaque assays for viral load, and flow cytometry. In WT mice placenta and uterine tissue expression of IL-10 was elevated with LCMV-Arm infection. Fetus resorption was elevated in WT on days 2 and 8 post-infection as compared to IL-10 KO, and by day 19 of gestation delivery was greater. Viral loads in the placenta and uterine tissue were resolved early in IL-10 KO mice, but persistent in tissues of WT mice. Levels of NRF2 and FAS were equivalent, but BCL2L11 was higher in IL-10 KO uterus. IL-6, Interferon-β (IFN-β), CCL2, and IL-17 levels were also equivalent. IL-10 KO tissues tended toward higher expression of interferon-γ (IFN-γ) and had significantly lower expression of Transforming growth factor beta (TGF-β). The proportion of placenta and uterine tissue CD8 T cells expressing the activation markers CD44 hi and PD1 were elevated in IL-10 KO mice. These data suggest that high IL-10 expression at the fetal-maternal interface following LCMV-Arm infection prevents clearance of viral load by impairing CD8 T cell activation and poses a significant threat to successful pregnancy outcome. The ability to modulate IL-10 expression at the maternal-fetal interface may help overcome negative pregnancy outcomes arising during acute LCMV and other viral infections in humans.
Article
We hypothesized that PD-1expressed by regulatory T cells (Tregs) would be functional and their expression levels may associate with activation status of CD4+ T and CD8+ T cells and the disease progression of HIV-1-infected patients. To prove it, we dynamically examined PD-1 expression levels by Tregs in peripheral blood of HIV-1-infected individuals not receiving antiretroviral therapy. Eighty-one HIV-1-infected individuals not undergoing antiretroviral therapy and 22 HIV-1-seronegative donors were enrolled in our study. Tregs were defined as CD4+CD25+CD127lo/- by flow cytometry. Expression of PD-1 and the activation markers CD38, HLA-DR, and Ki67 by Tregs and CD4+ T and CD8+ T cells was also determined by flow cytometry. TGF-β and IL-10 were measured to evaluate the suppressive function of Tregs. In all Tregs, the proportion of PD-1+ Tregs observed in HIV-1-infected persons was significantly greater than that seen in HIV-1-seronegative donors, and correlated with the activation of Tregs and CD4+ T and CD8+ T cells. This increased proportion of Tregs was also statistically associated with the disease progression. Blockade of PD-1/PD-L1 pathway with anti-PD-L1 mAb profoundly increased the level of intracellular TGF-β and IL-10 in CD4+CD25+CD127lo/- Tregs. Our data not only support that PD-1 plays a critical role to predict the activation status of cellular immunity and disease progression during HIV-1 infection but also indicate that blockade of PD-1/PD-L1 pathway represents a novel therapeutic approach to AIDS.
Article
Background: The lymphocyte-monocyte ratio (LMR) in the peripheral blood is suggested to be a potential biomarker for predicting the clinical outcomes of several diseases. We aimed to evaluate the relative efficiency of LMR for predicting 3-month mortality in patients with acute-on-chronic liver failure (AoCLF). Patients and methods: In this study, 74 chronic hepatitis B patients, 90 AoCLF patients, and 70 healthy controls were followed up for 4 months. The primary endpoint was 3-month in-hospital mortality. Hematological and virological parameters as well as liver biochemistry were determined using blood samples ordered upon admission. A panel of clinical and biochemical variables were analyzed for potential associations with outcomes using Cox proportional hazards and multiple regression models. Results: A significantly lower LMR was detected in AoCLF patients than in healthy controls and chronic hepatitis B groups (both P=0.001). The LMR inversely correlated with model for end-stage liver disease scores, and a lower LMR was associated with increased 3-month mortality. Multivariate analysis suggested that both LMR and model for end-stage liver disease scores were independent predictors of 3-month mortality (P<0.01). Conclusion: A low LMR measured at admission is predictive of a poor prognosis in AoCLF patients.
Article
Full-text available
The classical type of programmed cell death is characterized by its dependence on de novo RNA and protein synthesis and morphological features of apoptosis. We confirmed that stimulated 2B4.11 (a murine T-cell hybridoma) and interleukin-3 (IL-3)-deprived LyD9 (a murine haematopoietic progenitor cell line) died by the classical type of programmed cell death. Assuming that common biochemical pathways might be involved in the deaths of 2B4.11 and LyD9, we isolated the PD-1 gene, a novel member of the immunoglobulin gene superfamily, by using subtractive hybridization technique. The predicted PD-1 protein has a variant form of the consensus sequence found in cytoplasmic tails of signal transducing polypeptides associated with immune recognition receptors. The PD-1 gene was activated in both stimulated 2B4.11 and IL-3-deprived LyD9 cells, but not in other death-induced cell lines that did not show the characteristic features of the classical programmed cell death. Expression of the PD-1 mRNA in mouse was restricted to the thymus and increased when thymocyte death was augmented by in vivo injection of anti-CD3 antibody. These results suggest that activation of the PD-1 gene may be involved in the classical type of programmed cell death.
Article
Full-text available
Experiments designed to distinguish virus-specific from non-virus-specific T cells showed that bystander T cells underwent apoptosis and substantial attrition in the wake of a strong T-cell response. Memory CD8 T cells (CD8+ CD44hi) were most affected. During acute viral infection, transgenic T cells that were clearly defined as non-virus specific decreased in number and showed an increase in apoptosis. Also, use of lymphocytic choriomeningitis virus (LCMV) carrier mice, which lack LCMV-specific T cells, showed a significant decline in non-virus-specific memory CD8 T cells that correlated to an increase in apoptosis in response to the proliferation of adoptively transferred virus-specific T cells. Attrition of T cells early during infection correlated with the alpha/beta interferon (IFN-α/β) peak, and the IFN inducer poly(I:C) caused apoptosis and attrition of CD8+CD44hi T cells in normal mice but not in IFN-α/β receptor-deficient mice. Apoptotic attrition of bystander T cells may make room for the antigen-specific expansion of T cells during infection and may, in part, account for the loss of T-cell memory that occurs when the host undergoes subsequent infections.
Article
Full-text available
Induction and maintenance of T cell memory is critical for the control of intracellular pathogens and tumors. Memory T cells seem to require few "maintenance signals," though often such studies are done in the absence of competing immune challenges. Conversely, although attrition of CD8(+) T cell memory has been characterized in heterologous viral models, this is not the case for bacterial infections. In this study, we demonstrate attrition of T cell responses to the intracellular pathogen Listeria monocytogenes (LM) following an immune challenge with a second intracellular bacterium, Mycobacterium bovis (bacillus Calmette-Guérin, BCG). Mice immunized with either LM or recombinant LM (expressing OVA; LM-OVA), develop a potent T cell memory response. This is reflected by peptide-specific CTL, IFN-gamma production, and frequency of IFN-gamma-secreting T cells to native or recombinant LM Ags. However, when the LM-infected mice are subsequently challenged with BCG, there is a marked reduction in the LM-specific T cell responses. These reductions are directly attributable to the effects on CD4(+) and CD8(+) T cells and the data are consistent with a loss of LM-specific T cells, not anergy. Attrition of the Ag (OVA)-specific T cell response is prevented when LM-OVA-immunized mice are challenged with a subsequent heterologous pathogen (BCG) expressing OVA, demonstrating memory T cell dependence on Ag. Although the reduction of the LM-specific T cell response did not impair protection against a subsequent LM rechallenge, for the first time, we show that T cell attrition can result in the reduction of Ag-specific antitumor (B16-OVA) immunity previously established with LM-OVA immunization.
Article
The immunoreceptor PD-1 is significantly up-regulated on exhausted CD8+ T cells during chronic viral infections such as HIV-1. However, it remains unknown whether PD-1 expression on CD8+ T cells differs between typical progressors (TPs) and long-term nonprogressors (LTNPs). In this report, we examined PD-1 expression on HIV-specific CD8+ T cells from 63 adults with chronic HIV infection. We found that LTNPs exhibited functional HIV-specific memory CD8+ T cells with markedly lower PD-1 expression. TPs, in contrast, showed significantly up-regulated PD-1 expression that was closely correlated with a reduction in CD4 T-cell number and an elevation in plasma viral load. Importantly, PD-1 up-regulation was also associated with reduced perforin and IFN- production, as well as decreased HIV-specific effector memory CD8+ T-cell proliferation in TPs but not LTNPs. Blocking PD-1/PD-L1 interactions efficiently restored HIV-specific CD8+ T-cell effector function and proliferation. Taken together, these findings confirm the hypothesis that high PD-1 up-regulation mediates HIV-specific CD8+ T-cell exhaustion. Blocking the PD-1/PD-L1 pathway may represent a new therapeutic option for this disease and provide more insight into immune pathogenesis in LTNPs.
Article
Using a variety of techniques, including limiting dilution assays (LDA), intracellular IFNγ assays, and Db-IgG1 MHC dimer staining to measure viral peptide-specific T cell number and function, we show here that heterologous virus infections quantitatively delete and qualitatively alter the memory pool of T cells specific to a previously encountered virus. We also show that a prior history of a virus infection can alter the hierarchy of the immunodominant peptide response to a second virus and that virus infections selectively reactivate memory T cells with distinct specificities to earlier viruses. These results are consistent with a model for the immune system that accommodates memory T cell populations for multiple pathogens over the course of a lifetime.
Article
An understanding of how T cell memory is maintained is crucial for the rational design of vaccines. Memory T cells were shown to persist indefinitely in major histocompatibility complex (MHC) class I–deficient mice and retained the ability to make rapid cytokine responses upon reencounter with antigen. In addition, memory CD8 T cells, unlike naı̈ve cells, divided without MHC–T cell receptor interactions. This “homeostatic” proliferation is likely to be important in maintaining memory T cell numbers in the periphery. Thus, after naı̈ve CD8 T cells differentiate into memory cells, they evolve an MHC class I–independent “life-style” and do not require further stimulation with specific or cross-reactive antigen for their maintenance.
Article
Using a variety of techniques, including limiting dilution assays (LDA), intracellular IFNgamma assays, and Db-IgG1 MHC dimer staining to measure viral peptide-specific T cell number and function, we show here that heterologous virus infections quantitatively delete and qualitatively alter the memory pool of T cells specific to a previously encountered virus. We also show that a prior history of a virus infection can alter the hierarchy of the immunodominant peptide response to a second virus and that virus infections selectively reactivate memory T cells with distinct specificities to earlier viruses. These results are consistent with a model for the immune system that accommodates memory T cell populations for multiple pathogens over the course of a lifetime.
Article
Transient T cell depletion occurs before the development of an effective immune response to infection. In this study we show that most T cells, regardless of specificity, are induced to express early activation markers soon after infection with Listeria monocytogenes or lymphocytic choriomeningitis virus. Ag-specific T cells are further activated to display late activation markers and undergo extensive proliferation. As Ag-specific T cells begin to expand, nonspecific T cells are depleted en masse and exhibit no sign of further activation or proliferation before their depletion. This selective depletion of nonspecific T cells is due to in situ death via apoptosis, as visualized by confocal microscopy. Thus, early activation and subsequent depletion of nonspecific T cells are integral parts of the immune response to proinflammatory infections. These results have important implications for our understanding of early events in the development of a robust T cell response.
Article
Virus-specific memory T cell populations demonstrate plasticity in antigen recognition and in their ability to accommodate new memory T cell populations. The degeneracy of T cell antigen recognition and the flexibility of diverse antigen-specific repertoires allow the host to respond to a multitude of pathogens while accommodating these numerous large memory pools in a finite immune system. These cross-reactive memory T cells can be employed in immune responses and mediate protective immunity, but they can also induce life-threatening immunopathology or impede transplantation tolerance and graft survival. Here we discuss examples of altered viral pathogenesis occurring as a consequence of heterologous T cell immunity and propose models for the maintenance of a dynamic pool of memory cells.