You are on page 1of 6

Control of Toxoplasma reactivation by rescue

of dysfunctional CD8+ T-cell response via


PD-1–PDL-1 blockade
Rajarshi Bhadraa, Jason P. Gigleya, Louis M. Weissb, and Imtiaz A. Khana,1
a
Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, DC 20037; and bDepartment of Medicine
and Pathology, The Albert Einstein College of Medicine, Bronx, NY 10461

Edited by Jitender P. Dubey, US Department of Agriculture, Beltsville, MD, and approved April 26, 2011 (received for review October 13, 2010)

In this study, we document that Toxoplasma gondii differentiation interaction with its receptor PDL-1 not only augmented poly-
and reactivation are mediated by systemic CD8 T-cell dysfunction functional CD8+ T-cell response, but it also inhibited parasite
during chronic infection. We demonstrate that CD8+ T-cell exhaus- recrudescence and prevented the mortality of chronically infec-
tion occurs despite control of parasitemia during early-chronic ted mice in a CD8+ T-cell–dependent manner. To the best of
toxoplasmosis. During later phases, these cells become exhausted, our knowledge, this report of parasite reactivation caused by
leading to parasite reactivation and mortality. Concomitant with PD-1 mediated CD8+ T-cell dysfunction is unique.
increased CD8+ T-cell apoptosis and decreased effector response,
this dysfunction is characterized by a graded elevation in expres-
Results
sion of inhibitory receptor PD-1 on these cells in both lymphoid Toxoplasma-Infected Mice Show Increased Tachyzoite-Specific Gene
and nonlymphoid tissue. Blockade of the PD-1–PDL-1 pathway Expression with Concomitant Decrease in CD8+ T-Cell Response During
reinvigorates this suboptimal CD8+ T-cell response, resulting in Chronic Toxoplasmosis. Humans are most commonly infected
control of parasite reactivation and prevention of mortality in orally with Type II strains of Toxoplasma (11). To establish the
chronically infected animals. To the best of our knowledge, this loss of immune control during chronic infection, we first wanted
report is unique in showing that exposure to a persistent patho- to indentify the kinetics of long-term survival in B6 mice infected
gen despite initial control of parasitemia can lead to CD8+ T-cell orally with type II strain (ME49) cysts. Most of these animals
dysfunction and parasite reactivation. succumb to infection after 7 wk of challenge (Fig. 1A). Host
innate and adaptive immunity are thought to cause differentia-
tion of tachyzoites into chronic slow growing bradyzoites (12).
recrudescence | Toxoplasma gondii | adaptive immunity Because encysted parasites can persist for life and are not be-
lieved to be associated with disease pathology, several studies
Downloaded from https://www.pnas.org by 154.160.0.143 on February 15, 2024 from IP address 154.160.0.143.

T oxoplasmic encephalitis (TE) from reactivation of chronic


Toxoplasma infection affects 24% to 47% of HIV-infected
Toxoplasma-seropositive patients in the United States (1–3). Al-
have postulated that rather than an incidence of parasites, which
remain primarily encysted in chronically infected brains, the
degree of bradyzoite-tachyzoite interconversion is a better hall-
though the incidence of TE among AIDS patients declined mark of disease reactivation (6, 13, 14). Therefore, we assayed
threefold in the United States because of prophylactic treatment parasite reactivation by changes in parasite stage-specific gene
and highly active antiretroviral therapy, it still remains a signifi- expression. We measured the transcription of the tachyzoite
cant problem in developing countries (4, 5). Hence, understanding stage-specific Enolase-II (ENO-2) and the bradyzoite stage-spe-
the mechanisms responsible for parasite reactivation in chroni- cific Enolase-I (ENO-1), two genes that have been previously
cally infected hosts is critical for evolving strategies for the de- shown to change in very high levels (15–17). Concomitant with
velopment of immunotherapeutic agents against the parasite. poor survival of Toxoplasma gondii-infected mice during the late-
In the intermediate host, Toxoplasma undergoes stage conver- chronic phase, increased ENO-2 expression and decreased
sion between the rapidly replicating tachyzoite that is thought ENO-1 expression was noted at week 7 postinfection, suggesting
to be responsible for acute toxoplasmosis and the slowly repli- parasite reactivation (Fig. 1B). Interestingly, we observed that
cating, relatively quiescent, primarily encysted bradyzoite stage expression of both ENO-2 and ENO-1 occurs at week 10, in-
that can persist possibly for life (6). Effective CD8+ T-cell re-
dicating that despite limited tachyzoite to bradyzoite differenti-
sponse, critical for control of both acute and chronic Toxoplas-
ation, the parasites are still undergoing reactivation (i.e., a
mosis in susceptible mice strains paradoxically do not ensure their
bradyzoite to tachyzoite conversion) (Fig. 1B). To verify that
long-term survival (7). Poor long-term survival of Toxoplasma-
ENO-2 and ENO-1 corresponded to the tachyzoite and brady-
infected B10 mice is associated with reduced number of in-
tracerebral T cells and TE development (8). Additionally, de- zoite stage of T. gondii, the gene expression of the parasite were
pletion of CD8+ T cells rather than CD4+ T cells had a greater examined again in vivo using established models of acute and
effect on mortality of chronically infected animals (9). Taken to- chronic infection. Mice were infected with Type I RH strain
gether, these studies suggest that although critical for preventing (lethal acute infection) or Type II ME49 strain (chronic) (18,
parasite reactivation, CD8+ T-cell responses during late-chronic 19). Some of the ME49-infected animals were treated with drugs
infection are incapable of preventing mortality in infected mice. known to control reactivation of chronic infection (18, 19). As
However, the mechanism underlying this apparently ineffective expected, RH-infected mice exhibited high tachyzoite-specific
CD8+ T-cell response during late-chronic infection has not been gene expression (SAG-1, ENO-2) and minimal bradyzoite-spe-
studied and it remains to be determined if this defect is responsible
for loss of parasite control. We therefore tested whether CD8+ T
cells during late-chronic toxoplasmosis become dysfunctional, as Author contributions: R.B., J.P.G., and I.A.K. designed research; R.B., J.P.G., and L.M.W.
reported in some viral infections (10). performed research; L.M.W. contributed new reagents/analytic tools; R.B., J.P.G., L.M.W.,
and I.A.K. analyzed data; and R.B., J.P.G., L.M.W., and I.A.K. wrote the paper.
This article is unique in demonstrating that CD8+ T cells
during later phases of chronic toxoplasmosis are unable to mount The authors declare no conflict of interest.

a potent anamnestic response, a hallmark of robust adaptive This article is a PNAS Direct Submission.
immunity. Moreover, CD8+ T cells from these mice exhibit a 1
To whom correspondence should be addressed. E-mail: mtmixk@gwumc.edu.
progressive increase in expression of inhibitory molecule PD-1 This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.
with concomitant parasite reactivation. In vivo blockade of PD-1 1073/pnas.1015298108/-/DCSupplemental.

9196–9201 | PNAS | May 31, 2011 | vol. 108 | no. 22 www.pnas.org/cgi/doi/10.1073/pnas.1015298108


A 100 stage-specificity of the above markers using Type II parasites

Percent Survival
80
maintained in vitro under tachyzoite- or bradyzoite-inducing
60
conditions (20). Immunofluorescent microscopic analysis showed
40
20
that stage-specific protein expression corresponded with our
0 stage-specific transcript profile (Fig. S1).
0 50 100 150
Days post-infection Tissue cysts in brain occur almost exclusively in neurons and not
in leukocytes during the chronic phase (21). Because a recent
B Relative Expression 1.5
p=0.0006
ENO-2
2.5
ENO-1
study has shown that leukocytes can be used for parasite dissem-
0.8
p=0.025
2.0 p=0.0135 ination during the acute phase (22), we examined these cells for
0.6 1.5 parasitization as a further readout of reactivation. To character-
0.4 1.0 ize, whether parasite reactivation increased the frequency of
0.2 0.5 Toxoplasma-infected leukocytes, mononuclear leukocyte-gated
3 5 7 10 3 5 7 10
Weeks post-infection blood and brain cells were examined for parasites by flow cytom-
etry. Although minimal infection of blood and brain leukocytes
C Early Non-Moribund Moribund
Chronic Late chronic Late chronic
was noted during the early-chronic phase, high infection rates
Week 4-5 Week 7-9 Week 7-13 were observed during the late-chronic period, particularly in
39.1 0.0 32.8 0.1 17.7 0.2
moribund mice (Fig. 1C). Similar to observations noted by Courret
Brain

et al. (22), the level of parasite infection was much higher in cir-
0.4 7.3 25.8
culating leukocytes vis-à-vis brain leukocytes assayed at the same
CD8

27.3 0.0 12.0 0.0 9.9 0.1


time. It has been speculated that this may be because of multiple
Blood

populations of nonleukocytic brain lineages that are differentially


6.4 29.1 42.0
T. gondii permissive to T. gondii in the brain (22). Next, we assessed if
Spleen Brain reactivated parasites preferentially infected specific leukocyte
D 12.0 5.0 subsets in a site-dependent manner. Irrespective of tissue, the
Absolute Number
of CD8 T cells

majority of parasitized leukocytes corresponded to a myeloid


phenotype (CD3CD19NK1.1−CD11bhiF4/80+GR1+) (Fig. S2).
4
6
x 10

x 10

9.0 2.5
+
Taken together, our data suggest that during the late-chronic
6.0 0.0 phase, parasite-stage conversion results in reactivation of disease.
0 3 5 7 10 0 3 5 7 10
Weeks post-infection As Toxoplasma infection induces a robust CD8+ T-cell im-
munity that is known to be critical for long-term protection, it is
E Gating Strategy enigmatic that this potent immune response failed to prevent
recrudescence of infection (9). By measuring the kinetics of
CD90.1

CD8+ T-cell immunity we demonstrate that CD8+ T-cell re-

IMMUNOLOGY
sponse peaked at weeks 3 to 5 postinfection followed by gradual
Downloaded from https://www.pnas.org by 154.160.0.143 on February 15, 2024 from IP address 154.160.0.143.

CD8
-
Gated on CD8+CD90.1 T cells contraction (Fig. 1D). Concomitant with this process, reduced
Naive Week 5 Week 7
0.7 0.0 4.8 11.4 4.1 6.2 frequency of splenic and brain IFN-γ+Granzyme B+ and IFN-
γ−Granzyme B+ CD8+ T cells was noted (Fig. 1E).
Spleen

0.5 43.8 31.5


One of the quintessential features of robust long-term CD8+
T-cell immunity is the generation of a rapid response upon
IFNγ

0.9 4.2
3.5 1.7
secondary challenge (23). To verify the potency of secondary
Brain

36.6 17.0
response, Toxoplasma-infected animals were rechallenged during
Granzyme B
the early-chronic and late-chronic phases and assessed for cy-
cling CD8+ T cells (Fig. 1F). Mice rechallenged during the early-
chronic phase demonstrated a significant increase in absolute
CD8 Ki-67 Splenic T cells

F 2.5 p=0.0031
number of proliferating splenic CD8+ T cells over unchallenged
Absolute Number of

2.0 mice (Fig. 1F). In contrast, no significant change in cycling CD8+


x 10 6

+ 1.5 T cells was noted in rechallenged animals during the late-chronic


phase. These data clearly demonstrate that a potent recall re-
+ 1.0
Week 5 Week 7 sponse, a hallmark of robust CD8+ T-cell immunity, is com-
No Rechallenge Rechallenge
promised during later stages of chronic toxoplasmosis.
Fig. 1. Parasite reactivation during late-chronic T. gondii infection corre-
lates with poor CD8+ T-cell response. (A) C57BL/6 mice were infected with 10 CD8+ T Cells Up-Regulate PD-1 During Chronic Toxoplasmosis. The
ME49 cysts orally and survival was monitored on a daily basis. (B) Relative role of PD-1 in mediating dysfunction in chronic parasitic models
expression of ENO-1 and ENO-2 at weeks 3, 5, 7, and 10 postinfection in the has not been extensively addressed (24, 25). Although PD-1 ex-
brain was computed for each sample. Transcript levels at day 10 post- pression during toxoplasmosis has been recently reported (26),
inifection was taken as 1. (C) T. gondii-infected cells in the brain and blood its role in mediating CD8+ T-cell dysfunction or pathogenesis as
leukocyte-gated samples were assayed by flow cytometry. (D) The absolute shown in chronic viral models like lymphocytic choriomeningitis
number of CD8+ T cells at weeks 3, 5, 7, and 10 postinfection. (E) IFN-γ and virus (LCMV), hepatitis B virus (HBV), hepatitis C virus (HCV),
Granzyme B production by CD8+ T cells was evaluated in splenocytes or brain and HIV (27), is currently unknown. Analysis of CD8+ T cells
cells from infected mice in presence Toxoplasma lysate antigen. (F) At weeks revealed a progressive increase of PD-1 expression and fre-
5 and 7 after primary infection, mice were rechallenged orally with 10 cysts quency during the chronic stage of parasite infection (Fig. 2). Of
and animals were killed 4 d later. The absolute number of cycling CD8+ the two PD-1 ligands, PDL-1 and PDL-2, the mean fluorescence
T cells was evaluated in the spleen during early-chronic and late-chronic intensity (MFI) of the former but not the latter increased during
infection with or without rechallenge. The data represent two experiments
the course of chronic infection (Fig. S3).
with at least four mice per group. Error bars represent SD throughout.
Because increased PD-1 MFI correlated with decreased fre-
quency of splenic IFN-γ+CD8+ T cells, we hypothesized that
PD-1hiCD8+ T cells would have reduced capacity to release
cific gene expression (BAG-1, ENO-1) (Fig. S1). In contrast, IFN-γ. To address this theory, we measured the proportion of
drug-treated mice showed reduced SAG-1 and ENO-2 transcript IFN-γ–producing cells within PD-1hiCD8+ and PD-1intCD8+ T-
levels and increased BAG-1 and ENO-1 expression vis-à-vis un- cell population. No significant difference in cytokine-positive
treated ME49 infected mice. Next, we further confirmed the cells between the two populations was noted (Fig. S4). Although

Bhadra et al. PNAS | May 31, 2011 | vol. 108 | no. 22 | 9197
Spleen Liver Brain be attributed to attenuated inflammation or antigen load in drug-
1.3 0.7
treated mice, which can result in suboptimal development or

Naive
maintenance of Granzyme B-expressing short-lived effector sub-
10.5 2.2 14.2 set (31, 32). Combined, these data suggest that control of infection

Week 10 Week 7 Week 5 Week 3


% of maximum
during the early phases of infection may be critical for minimizing
10.2 2.7 11.4 induction of CD8 dysfunction during the chronic phase.

Blockade of PD-1–PDL-1 Pathway Reinvigorates CD8+ T-Cell Response.


22.4 4.3 23.5
Next, we addressed if a dysfunctional CD8+ T-cell response
could be revived by blocking the PD-1–PDL-1 pathway. To ad-
20.4 4.3 29.5 dress this, animals were administered a nondepleting anti–PDL-
1 antibody starting at week 5 postinfection for a period of 2 wk
PD-1 and, thereafter, the CD8+ T-cell response in the spleen, liver,
and brain were quantified (33). Although significant increase in
Fig. 2. PD-1 is expressed at high levels on CD8+ T cells in both lymphoid and absolute number of CD8+ T cells was noted in all of the tissues
nonlymphoid tissue during late-chronic Toxoplasmosis. PD-1 expression on examined, the most dramatic increase occurred in brain (Fig.
CD8+ T cells from tissues (spleen, brain, and liver) was measured at weeks 3,
3A). To determine if this increase was because of enhanced
5, 7, and 10 postinfection. Filled histogram represents staining with isotype-
proliferation, we measured Ki-67 expression in the CD8+ T cells.
control antibody. The data represent three experiments with at least four
mice per group.
Unlike the spleen and liver, we were unable to detect increased
frequency of cycling CD8+ T cells in anti–PDL-1 treated groups
in the brain (Fig. 3B). Taken together, these data suggest that
this finding suggests that PD-1 expression does not directly one of the mechanisms for increased brain CD8+ T cells in anti–
regulate IFN-γ production in CD8+ T cells, the role of other PDL-1 treated mice is not in situ proliferation but possibly in-
factors, such as the extent and duration of PD-1 cross-linking and creased recruitment of these cells to the tissue.
frequency of PD-1–PDL-1 engagement in regulating CD8+ T- As IFN-γ production (34) and cytotoxicity (35) are known to
cell–mediated IFN-γ production, cannot be ruled out. Irre- be critical for control of toxoplasmosis, we examined if blockade
spective of the time point, very few PD-1lo cells produced IFN-γ, with anti–PDL-1 augmented these functions within the CD8+ T-
indicating that an overwhelming majority of CD8+ T cells in this cell population. Anti–PDL-1 treatment increased the frequency
of IFN-γ+Granzyme B+ and IFN-γ−Granzyme B+ CD8+ T cells
subset are naive. Although we did not observe a significant dif-
in the spleen and brain, as well as ex vivo cytotoxicity of splenic
ference in frequency of IFN-γ+CD8+ T cells between the PD-1hi
CD8+ T cells (Fig. 3 C and D). We then measured, if anti–PDL-1
and PD-1int CD8+ T-cell subsets, we hypothesized that the re-
treatment decreased the frequency of PD-1hi cells. Surprisingly,
duced frequency of splenic IFN-γ+CD8+ T cells during late-
PD-1–PDL-1 blockade caused a further increase in the fre-
chronic infection could be because of apoptosis of PD-1hi cells. As quency of PD-1–expressing CD8+ T cells (Fig. 3E). Because we
Downloaded from https://www.pnas.org by 154.160.0.143 on February 15, 2024 from IP address 154.160.0.143.

recent studies have suggested that apoptosis can be coupled to demonstrated that high PD-1 expression was associated with
cell-cycle progression (28, 29), we incubated splenocytes from apoptosis, we determined if rescue of CD8+ T-cell response by
week 7-infected mice for 5 h at 37 °C and stained for CD8, PD-1, PD-1–PDL-1 blockade was because of reduced apoptosis. To
Ki-67, and active caspase-3, the active form of the apoptosis-re- address this under steady-state conditions without confounding
lated cysteine peptidase. Although cycling PD-1hiCD8+ T cells effects of proliferation and graded PD-1 increase, we evaluated
exhibited a higher expression of active caspase-3 than noncycling apoptotic CD8+ T cells in splenocytes from chronically infected
PD-1hiCD8+ T cells, PD-1int/lo cells had a much lower MFI than mice incubated for 5 h with anti–PDL-1 or control antibody (Fig.
cycling or noncycling PD-1hi cells (Fig. S4), suggesting that cycling S7). Anti–PDL-1 treatment mediated reduction in apoptosis
PD-1hiCD8+ T cells exhibit poor survival. occurred primarily in the PD-1int subset and only moderate de-
Because PD-1 has been reported to be transiently expressed in crease in apoptosis was noted in the PD-1hi subset. To further
acute viral infections (27), increased PD-1 MFI during later address if anti–PDL-1 treatment was capable of rescuing pre-
stages of chronic Toxoplasmosis could be because of a genera- existing PD-1 expressing CD8+ T cells in vivo, we adoptively
tion of new PD-1–expressing effectors in response to parasite transferred sorted PD-1+CD8+ T cells from chronically infected
recrudescence. To address this theory, we used a BrdU pulse- CD45.1 mice (week 5–6) to similarly infected CD45.2 mice and
chase approach and then measured PD-1 expression on BrdU+ then treated recipients with anti–PDL-1 antibody. Anti–PDL-1
and Brdu− CD8+ T cells during the early-chronic and late-chronic treatment increased the absolute number of donor cells in both
phases (Fig. S5). Dramatic increase in PD-1 MFI and frequency of spleen and brain (Fig. S8). However, although the recipient
PD-1hi cells was noted only in BrdU-labeled fraction. This finding CD8+ T-cell population (comprising of both PD-1+ and PD-1−)
suggests that PD-1 up-regulation is a graded process and the expanded sixfold at the effector site (vis-à-vis isotype-treated),
majority of PD-1hiCD8+ T cells at week 7 postinfection are not the donor population underwent over 20-fold expansion. This
new effector cells generated in response to parasite recrudescence. finding suggests that anti–PDL-1 treatment preferentially rescues
Considering that a majority of PD-1hiCD8+ T cells during the preexisting PD-1–expressing cells.
late-chronic phase are not newly generated effectors, we wanted to
address if the control of infection during the early or later phases of Blockade of the PD-1–PDL-1 Pathway Controls Toxoplasma Recrudescence.
infection would result in reduced CD8 exhaustion. To evaluate Finally, we wanted to address if this augmented CD8+ T-cell re-
this, we treated mice with sulfamethoxazole and trimethoprim at sponse was able to control recrudescence by examining the gene
various time points postinfection, as show in Fig. S6 (19). Because expression of Toxoplasma parasites in brains of anti–PDL-1 or
reduced antigen burden in drug-treated mice can result in in- control antibody treated mice. Anti–PDL-1 treated mice revealed
creased naive T-cell numbers, which can potentially confound more of a bradyzoite-specific than tachyzoite-specific gene ex-
interpretation of data, we analyzed brain, a tissue known to recruit pression (Fig. 4A). Moreover, anti–PDL-1 treated mice exhibited
CD8+ T cells in a strictly antigen-specific manner (30). Only early a much lower frequency of Toxoplasma-infected leukocytes in
initiation of drug treatment resulted in decreased PD-1 MFI on brain and blood than control group (Fig. 4B). As previously stated,
CD8+ T cells during the late-chronic phase (Fig. S6). Moreover, because encysted parasites can persist for life in immunocompe-
CD8+ T cells from these mice exhibited a higher proportion of tent host brains without causing pathology, we examined parasite
IFN-γ+GranzymeB− and IFN-γ+GranzymeB+ CD8+ T cells (Fig. genomic DNA in blood (6). Detection of parasite DNA in blood
S6). However, IFN-γ−GranzymeB+ CD8+ T cells underwent by PCR has been used to diagnose cerebral toxoplasmosis in
a moderate decrease in these animals (Fig. S6). This finding may patents with AIDS (36). As shown in Fig. 4C, parasite genomic

9198 | www.pnas.org/cgi/doi/10.1073/pnas.1015298108 Bhadra et al.


A 9.0
p=.0001 increased survival of anti–PDL-1 treated mice was CD8-dependent,
the surviving mice were treated with anti-CD8 antibody. As shown

Absolute Number of CD8 T cells


6
Spleen

x 10
6.0 in Fig. 4E, CD8 depletion abrogated the survival of anti–PDL-1
+
3.0 treated animals. Taken together, the above data suggest that anti–
5.0 p=.0002
PDL-1 treatment augments suboptimal CD8+ T-cell response in

5
chronically infected mice, which prevents reactivation of infection

Liver

x 10
3.5
leading to host survival.
2.0
p=.0006
1.1
Anti–PDL-1 Treatment Up-Regulates Eomes in CD8+ T Cells from
Brain

4
x 10
0.6
Chronically Infected Mice. T-box factors, T-bet and Eomeso-
0.1 dermin, play a critical role in development, survival, and function
Isotype Anti-
Control PDL-1
of CD8+ T cells (37, 38). Studies from our laboratory suggest that
anti–PDL-1 treatment dramatically augments expression of
Isotype Control Anti-PDL-1 Eomes but not T-bet (unaffected vis-à-vis acutely infected mice) in
B lo int hi lo int hi cycling CD8+ T cells (Fig. S9). Incidentally, the critical role of
Eomes in mediating CD8+ T-cell responsiveness to IL-15 (38),
Spleen

14.8 15.1 17.9 22.9 a cytokine crucial for survival of memory as well as effector CD8+
T cells, and Granzyme B (39–41) expression in T cells has been
recently elucidated. Significantly, our data show that CD8+ T-cell
Liver
CD8

26.0 6.5 30.0 10.1


dysfunction affects Granzyme B expression more than IFN-γ
production. Significance of Eomes expression in our current
Brain

28.6 3.2 25.2 4.4


model will have to await further investigation.
Ki-67 Discussion
-
Gated on CD8+CD90.1 T cells
CD8+ T-cell exhaustion has been reported in several chronic
C Isotype Control Anti-PDL-1 viral infections, like LCMV, HBV, HCV, HIV, and Simian Im-
5.2 4.8 4.1 9.1 munodeficiency Virus, which are characterized by high levels of
Spleen

persisting viremia (42). This exhaustion is manifested by the


gradual loss of CD8+ T-cell effector functions (cytokine pro-
18.6 29.2 duction, cytotoxicity, proliferation, and recall responses) and, in
IFNγ

5.3 2.6 6.7 6.0 extreme situations, CD8+ T cells can be physically deleted (43).
In contrast, in infection models like murine cytomegalovirus,
Brain

characterized by low levels of persistent viremia, antigen-specific

IMMUNOLOGY
23.0 33.5 T cells remain functional and respond vigorously to viral chal-
Downloaded from https://www.pnas.org by 154.160.0.143 on February 15, 2024 from IP address 154.160.0.143.

Granzyme B lenge (42, 44). In models of reactivation like HSV that induce
latent infection punctuated by periods of reactivation, functional
D 60 Infected + Isotype Control long-lived CD8+ T-cell response is generated (45). Based on
Infected + Anti-PDL-1
these findings, it has been suggested that CD8+ T-cell exhaustion
Percent Lysis

40 Naive
occurs only in the presence of uncontrolled persistent viremia
20 (42, 46–48). Our current report documents that CD8+ T-cell
dysfunction is not restricted to viral infections alone but can be
0
10:1 20:1 40:1 extended to intracellular protozoan parasite, like T. gondii. Al-
Effector:Target Ratio though CD8+ T-cell dysfunction observed in the present study is
similar in certain facets to certain viral pathogens, in its de-
E Isotype Control pendence on PD-1–PDL-1 interaction, it discounts the conven-
% of maximum

Anti-PDL-1
tional paradigm that CD8+ T-cell exhaustion occurs only in
infectious disease models with uncontrolled persistent pathogen
load. Moreover, notwithstanding the fact that the role of the
PD-1–PDL-1 pathway has been addressed in chronic viral
PD-1 models of CD8+ T-cell exhaustion, its significance in bacterial
or parasitic models of CD8+ T-cell dysfunction has not been
Fig. 3. Blockade of PD-1–PDL-1 pathway reinvigorates CD8+ T-cell response extensively investigated.
in both lymphoid and nonlymphoid tissue in chronically infected mice. (A) Our studies demonstrate that despite a potent development of
Chronically infected mice were treated with anti–PDL-1 for 2 wk and then CD8+ T-cell immunity during the early stages of chronic infection,
killed at week 7 postinfection. The absolute number of CD8+ T cells was
endogenous CD8+ T-cell response is unable to control parasite
evaluated in the spleen, liver, and brain. (B) The frequency of proliferating
CD8+ T cells was assessed by intracellular staining for Ki-67. (C) IFN-γ and
reactivation at the later time points. This finding is in agreement
Granzyme B production by CD8+ T cells was evaluated in splenocytes or brain
with a previous study by Gazzinelli et al. (49), which demonstrated
cells from infected mice in the presence of TLA. (D) Antigen-specific cyto- that parasite reactivation in B6 mice during late-chronic phases is
toxicity of splenic CD8+ T-cells was measured by 51Cr-release assay. (E) PD-1 associated with increased expression of tachyzoite-specific genes
expression on splenic CD8+ T cells from anti–PDL-1 or isotype control anti- in brain. Additionally, there is an increase in parasite levels in the
body-treated mice was evaluated by flow cytometry. The data represent periphery. A recent study has demonstrated the CD11b+ leuko-
three experiments with at least four mice per group. cytes can act as shuttles for parasite migration during the acute
infection phase (22). Hence, it would be interesting to investigate
in future studies whether during parasite reactivation, CD11b+
leukocytes, which are preferentially parasitized during the late-
DNA was only detectable in control antibody-treated and not in
chronic phase, potentially serve a similar function and its role in
drug or anti–PDL-1 treated blood. Next, we monitored long-term mediating disease pathogenesis. Cumulatively, our results show
survival of anti–PDL-1 treated mice. Surprisingly, although 100% that long-term exposure to a persistent pathogen despite initial
of control mice died by day 91 postinfection, 90% of anti–PDL-1 control of parasitemia can lead to CD8+ T-cell dysfunction and
treated mice survived until the termination (day 125 postinfection) parasite reactivation. This CD8 exhaustion with concomitant PD-
of the experiment (Fig. 4D). Furthermore, to demonstrate that 1 up-regulation is characterized by reduced effector-cell numbers,

Bhadra et al. PNAS | May 31, 2011 | vol. 108 | no. 22 | 9199
A ENO-2 ENO-1 dysfunction. This finding suggests that high antigen burden or

Relative Expression
1.5 p=0.012 2.0 p=0.038 inflammation (including anti-inflammatory cytokines) or both that
1.5
1.0 occurs during acute phase of infection may be primarily re-
1.0
0.5
sponsible for defective development of a long-lived robust CD8
0.5
response during the chronic phase (42, 50). Based on a recent
0.0 0.0
Isotype Control
study by Chang et al. (32) demonstrating that T-cell fate im-
Anti-PDL-1 printing can occur as early as the first cell division, it is hardly
surprising that T-cell microenvironment (cytokine, antigen bur-
B Isotype Control Anti-PDL-1
26.7 0.2 41.1 0.1
den, and so forth) during the early phase of Toxoplasma infection
can have such a profound effect during the later phases. Our
Brain

findings are further supported by a study in an LCMV model


16.0 4.5
CD8

11.3 0.3 25.2 0.0


where antigen burden has been shown to be the key determinant
for development of CD8 dysfunction (42). However, any control
Blood

of infection or antigen burden can potentially affect inflammation.


36.5 3.9
T. gondii
Hence, in future studies it will be critical to discriminate the dif-
ferential role of inflammatory milieu and antigen burden in me-
diating CD8 dysfunction.
per 400ng of whole blood

C 30
Recent reports have identified role of IL-10 in mediating
Parasite DNA levels

20 dysfunctional CD8+ T-cell response during LCMV infection


(51). However, anti-IL10R treatment of mice carrying chronic
10 Toxoplasma infection led to their increased mortality because of
0
a strong inflammatory response (52). This finding suggests that
any strategy for preventing Toxoplasma reactivation must involve
Treatment

Control

PDL-1
Isotype

Treated
Anti-
Drug

an appropriate balance between enhanced proinflammatory


Pre

mediators without excessive suppression of anti-inflammatory


Week 5 Week 7 factors. Interestingly, anti–PDL-1 treatment not only augmented
the CD8+ T-cell response but also prevented mortality in
D Anti-PDL-1 treatment
chronically infected mice by controlling parasite reactivation.
How blockade of the PD-1–PDL-1 pathway modulates CD8+
0 35 49
Days post-infection
125 T-cell response at the molecular level is poorly understood in
100 infectious disease models. Recent studies have shown that dif-
ferential TCR signal strength can affect not only memory CD8
Percent Survival

80
p<0.0001
programming but also development of a potent polyfunctional
60
CD8 response (53, 54). Considering that PD-1 ligation has been
Downloaded from https://www.pnas.org by 154.160.0.143 on February 15, 2024 from IP address 154.160.0.143.

40
shown to inhibit TCR signaling via down-regulation of ZAP-70
20 Anti-PDL-1 and PKCθ in vitro, it is possible that anti–PDL-1 treatment
Isotype Control
0 restores robust CD8 response during chronic Toxoplasma in-
0 28 56 84 125
Days post-infection fection via increased TCR sensitivity (55). Although the mech-
anistic basis of CD8 dysfunction and PD-1 up-regulation need
E Anti-CD8 Anti-PDL-1 to be investigated more thoroughly in future studies, the data
treatment treatment presented in this article provide significant insights into the role
of PD-1–PDL-1 pathway in mediating CD8+ T-cell dysfunction
126 136 146 150
Days post-infection and concomitant parasite reactivation. These results are espe-
100 cially significant in view of a minimal role played by CTLA-4,
Anti-PDL-1 +
Percent Survival

80 Isotype Control a related inhibitory molecule in regulating T-cell response during


60 Anti-PDL-1 + Anti-CD8 TE (56). Our findings have important implications in the use
40
of PD-1–PDL-1 blockade in combination with drug regimen
20 for controlling TE in immunocompromised patients, as well as
p=.0067
0 use of similar therapy in other dysfunction-induced reactivation-
126 136 150
Days post-infection type diseases.
Fig. 4. Anti–PDL-1 treatment controls parasite reactivation and prevents
Materials and Methods
mortality in chronically infected mice. (A) Relative expression of ENO-1 and
ENO-2 was computed for each sample. Transcript levels at day 10 post- Mice, Parasites, and in Vivo Antibody Treatment. Female C57BL/6, CD45.1, and
infection was taken as 1. (B) T. gondii-infected cells in the brain and blood CD90.1 mice were purchased from the National Cancer Institute or the
leukocyte-gated samples (n = 5) were assayed by flow cytometry. (C) Parasite Jackson Laboratory. Animal studies were carried out in agreement with In-
genomic DNA level was computed in whole blood before initiation of stitutional Animal Care and Use Committee approved guidelines. Unless
treatment (week 5 postinfection) or 2 wk after initiation of treatment (week otherwise mentioned, T. gondii cysts of ME49 strain were used for i.g. in-
7 postinfection) (n = 4–5). (D) Anti–PDL-1 antibody was injected every third fection throughout. Some mice were injected in vivo with the following anti-
day, starting at week 5 postinfection (n = 10). To minimize the development bodies i.p.: anti-PDL-1 (MIH5) or anti-CD8 (2.43) or isotype-matched control
of an allogeneic response, the antibody was administered with 2-wk inter- antibodies.
ruptions until termination of the experiment. (E) Surviving anti–PDL-1
treated mice from the previous experiment were administered anti–PDL-1 Lymphocyte Isolation, Cell Surface Staining, and Intracellular Staining. Single
along with anti CD8 (n = 4) or anti–PDL-1 and control antibody (n = 4) every cell suspension was prepared from spleen, liver, blood, and brain using
third day unitl the conclusion of the experiment. Survival was monitored on standard protocol. For cytokine detection, 5 × 105 naïve congenic splenocytes
a daily basis. Data represent two experiments. was mixed with equal number of brain or splenic cells from infected mice,
and restimulation was similarly carried out in the presence of TLA.

Real-Time RT-PCR and PCR. From total infected mouse brain RNA, real-time RT-
decreased polyfunctionality, increased apoptosis, and poor recall PCR was performed with primers for ENO-1, ENO-2, SAG-1, and BAG-1 on
response and expansion. Preliminary studies using drug treatment Toxoplasma specific actin normalized samples. Transcript levels relative to
suggest that control of infection during the early but not later day 10 post infection (day 10 post infection relative transcript level = 1.0)
phases is critical for minimizing PD-1 up-regulation and CD8 were calculated according to the Pfaffl method of quantitation (57). Parasite

9200 | www.pnas.org/cgi/doi/10.1073/pnas.1015298108 Bhadra et al.


DNA quantitation (fg of DNA) was calculated after real-time PCR by com- Additional details and techniques are provided in SI Materials and
paring Ct values for the B1 gene in experimental samples to a serial dilution Methods.
of genomic parasite DNA equivalents as previously described (26).
ACKNOWLEDGMENTS. We thank M. Azuma for providing MIH5 hybridoma.
Statistical Analysis. Statistical analysis was performed used Student’s t test or This work was supported by National Institutes of Health Grant AI-33325
Log-rank test with P < 0.05 taken as statistically significant. (to I.A.K.).

1. Luft BJ, Remington JS (1992) Toxoplasmic encephalitis in AIDS. Clin Infect Dis 15: 28. Janumyan YM, et al. (2003) Bcl-xL/Bcl-2 coordinately regulates apoptosis, cell cycle
211–222. arrest and cell cycle entry. EMBO J 22:5459–5470.
2. Grant IH, Gold JW, Rosenblum M, Niedzwiecki D, Armstrong D (1990) Toxoplasma 29. Andersen JL, et al. (2006) HIV-1 Vpr-induced apoptosis is cell cycle dependent and
gondii serology in HIV-infected patients: The development of central nervous system requires Bax but not ANT. PLoS Pathog 2:e127.
toxoplasmosis in AIDS. AIDS 4:519–521. 30. Galea I, et al. (2007) An antigen-specific pathway for CD8 T cells across the blood-
3. Zangerle R, Allerberger F, Pohl P, Fritsch P, Dierich MP (1991) High risk of developing brain barrier. J Exp Med 204:2023–2030.
toxoplasmic encephalitis in AIDS patients seropositive to Toxoplasma gondii. Med 31. Joshi NS, et al. (2007) Inflammation directs memory precursor and short-lived effector
Microbiol Immunol (Berl) 180:59–66. CD8(+) T cell fates via the graded expression of T-bet transcription factor. Immunity
4. Jones JL, et al. (1999) Surveillance for AIDS-defining opportunistic illnesses, 1992– 27:281–295.
1997. MMWR CDC Surveill Summ 48:1–22. 32. Chang JT, et al. (2007) Asymmetric T lymphocyte division in the initiation of adaptive
5. Vidal JE, et al. (2005) Cerebral toxoplasmosis in HIV-positive patients in Brazil: Clinical immune responses. Science 315:1687–1691.
features and predictors of treatment response in the HAART era. AIDS Patient Care 33. Dias P, et al. (2010) CD4 T-cell help programs a change in CD8 T-cell function enabling
STDS 19:626–634. effective long-term control of murine gammaherpesvirus 68: Role of PD-1-PD-L1
6. Lyons RE, McLeod R, Roberts CW (2002) Toxoplasma gondii tachyzoite-bradyzoite interactions. J Virol 84:8241–8249.
interconversion. Trends Parasitol 18:198–201. 34. Suzuki Y, Orellana MA, Schreiber RD, Remington JS (1988) Interferon-gamma: The
7. Scharton-Kersten TM, Yap G, Magram J, Sher A (1997) Inducible nitric oxide is major mediator of resistance against Toxoplasma gondii. Science 240:516–518.
essential for host control of persistent but not acute infection with the intracellular 35. Hakim FT, et al. (1991) CD8+ T cells from mice vaccinated against Toxoplasma gondii
pathogen Toxoplasma gondii. J Exp Med 185:1261–1273. are cytotoxic for parasite-infected or antigen-pulsed host cells. J Immunol 147:
8. Deckert-Schlüter M, et al. (1994) Toxoplasma encephalitis in congenic B10 and BALB 2310–2316.
mice: Impact of genetic factors on the immune response. Infect Immun 62:221–228. 36. Lamoril J, et al. (1996) Detection by PCR of Toxoplasma gondii in blood in the
9. Gazzinelli R, Xu Y, Hieny S, Cheever A, Sher A (1992) Simultaneous depletion of CD4+ diagnosis of cerebral toxoplasmosis in patients with AIDS. J Clin Pathol 49:89–92.
and CD8+ T lymphocytes is required to reactivate chronic infection with Toxoplasma 37. Glimcher LH, Townsend MJ, Sullivan BM, Lord GM (2004) Recent developments in the
gondii. J Immunol 149:175–180. transcriptional regulation of cytolytic effector cells. Nat Rev Immunol 4:900–911.
10. Gallimore A, et al. (1998) Induction and exhaustion of lymphocytic choriomeningitis 38. Intlekofer AM, et al. (2005) Effector and memory CD8+ T cell fate coupled by T-bet
virus-specific cytotoxic T lymphocytes visualized using soluble tetrameric major and eomesodermin. Nat Immunol 6:1236–1244.
histocompatibility complex class I-peptide complexes. J Exp Med 187:1383–1393. 39. Pearce EL, et al. (2003) Control of effector CD8+ T cell function by the transcription
11. Su C, et al. (2003) Recent expansion of Toxoplasma through enhanced oral factor Eomesodermin. Science 302:1041–1043.
transmission. Science 299:414–416. 40. Cruz-Guilloty F, et al. (2009) Runx3 and T-box proteins cooperate to establish the
12. Hughes HP, Kasper LH, Little J, Dubey JP (1988) Absence of a role for natural killer transcriptional program of effector CTLs. J Exp Med 206:51–59.
cells in the control of acute infection by Toxoplasma gondii oocysts. Clin Exp Immunol 41. Sanjabi S, Mosaheb MM, Flavell RA (2009) Opposing effects of TGF-beta and IL-15
72:394–399. cytokines control the number of short-lived effector CD8+ T cells. Immunity 31:

IMMUNOLOGY
13. Ferguson DJ, Huskinson-Mark J, Araujo FG, Remington JS (1994) A morphological 131–144.
Downloaded from https://www.pnas.org by 154.160.0.143 on February 15, 2024 from IP address 154.160.0.143.

study of chronic cerebral toxoplasmosis in mice: Comparison of four different strains 42. Mueller SN, Ahmed R (2009) High antigen levels are the cause of T cell exhaustion
of Toxoplasma gondii. Parasitol Res 80:493–501. during chronic viral infection. Proc Natl Acad Sci USA 106:8623–8628.
14. Sibley LD, Khan A, Ajioka JW, Rosenthal BM (2009) Genetic diversity of Toxoplasma 43. Shin H, Wherry EJ (2007) CD8 T cell dysfunction during chronic viral infection. Curr
gondii in animals and humans. Philos Trans R Soc Lond B Biol Sci 364:2749–2761. Opin Immunol 19:408–415.
15. Fleige T, Fischer K, Ferguson DJ, Gross U, Bohne W (2007) Carbohydrate metabolism in 44. Snyder CM, et al. (2008) Memory inflation during chronic viral infection is maintained
the Toxoplasma gondii apicoplast: Localization of three glycolytic isoenzymes, the by continuous production of short-lived, functional T cells. Immunity 29:650–659.
single pyruvate dehydrogenase complex, and a plastid phosphate translocator. 45. Zhu J, et al. (2007) Virus-specific CD8+ T cells accumulate near sensory nerve endings
Eukaryot Cell 6:984–996. in genital skin during subclinical HSV-2 reactivation. J Exp Med 204:595–603.
16. Dzierszinski F, Mortuaire M, Dendouga N, Popescu O, Tomavo S (2001) Differential 46. Wherry EJ, Blattman JN, Ahmed R (2005) Low CD8 T-cell proliferative potential and
expression of two plant-like enolases with distinct enzymatic and antigenic properties high viral load limit the effectiveness of therapeutic vaccination. J Virol 79:8960–8968.
during stage conversion of the protozoan parasite Toxoplasma gondii. J Mol Biol 309: 47. Fuller MJ, Zajac AJ (2003) Ablation of CD8 and CD4 T cell responses by high viral loads.
1017–1027. J Immunol 170:477–486.
17. Fux B, et al. (2007) Toxoplasma gondii strains defective in oral transmission are also 48. Wherry EJ, Blattman JN, Murali-Krishna K, van der Most R, Ahmed R (2003) Viral
defective in developmental stage differentiation. Infect Immun 75:2580–2590. persistence alters CD8 T-cell immunodominance and tissue distribution and results in
18. Tait ED, et al. (2010) Virulence of Toxoplasma gondii is associated with distinct distinct stages of functional impairment. J Virol 77:4911–4927.
dendritic cell responses and reduced numbers of activated CD8+ T cells. J Immunol 49. Gazzinelli RT, Eltoum I, Wynn TA, Sher A (1993) Acute cerebral toxoplasmosis is
185:1502–1512. induced by in vivo neutralization of TNF-alpha and correlates with the down-
19. Torre D, et al.; Italian Collaborative Study Group (1998) Randomized trial of regulated expression of inducible nitric oxide synthase and other markers of
trimethoprim-sulfamethoxazole versus pyrimethamine-sulfadiazine for therapy of macrophage activation. J Immunol 151:3672–3681.
toxoplasmic encephalitis in patients with AIDS. Antimicrob Agents Chemother 42: 50. Mordue DG, Monroy F, La Regina M, Dinarello CA, Sibley LD (2001) Acute
1346–1349. toxoplasmosis leads to lethal overproduction of Th1 cytokines. J Immunol 167:
20. Weiss LM, et al. (1995) A cell culture system for study of the development of 4574–4584.
Toxoplasma gondii bradyzoites. J Eukaryot Microbiol 42:150–157. 51. Brooks DG, Lee AM, Elsaesser H, McGavern DB, Oldstone MB (2008) IL-10 blockade
21. Ferguson DJ, Hutchison WM (1987) The host-parasite relationship of Toxoplasma facilitates DNA vaccine-induced T cell responses and enhances clearance of persistent
gondii in the brains of chronically infected mice. Virchows Arch A Pathol Anat virus infection. J Exp Med 205:533–541.
Histopathol 411:39–43. 52. Jankovic D, et al. (2007) Conventional T-bet(+)Foxp3(-) Th1 cells are the major source
22. Courret N, et al. (2006) CD11c- and CD11b-expressing mouse leukocytes transport of host-protective regulatory IL-10 during intracellular protozoan infection. J Exp
single Toxoplasma gondii tachyzoites to the brain. Blood 107:309–316. Med 204:273–283.
23. Zammit DJ, Cauley LS, Pham QM, Lefrançois L (2005) Dendritic cells maximize the 53. Teixeiro E, et al. (2009) Different T cell receptor signals determine CD8+ memory
memory CD8 T cell response to infection. Immunity 22:561–570. versus effector development. Science 323:502–505.
24. Langhorne J, Ndungu FM, Sponaas AM, Marsh K (2008) Immunity to malaria: More 54. Almeida JR, et al. (2009) Antigen sensitivity is a major determinant of CD8+ T-cell
questions than answers. Nat Immunol 9:725–732. polyfunctionality and HIV-suppressive activity. Blood 113:6351–6360.
25. Bustamante JM, Bixby LM, Tarleton RL (2008) Drug-induced cure drives conversion to 55. Sheppard KA, et al. (2004) PD-1 inhibits T-cell receptor induced phosphorylation of
a stable and protective CD8+ T central memory response in chronic Chagas disease. the ZAP70/CD3zeta signalosome and downstream signaling to PKCtheta. FEBS Lett
Nat Med 14:542–550. 574:37–41.
26. Wilson EH, et al. (2009) Behavior of parasite-specific effector CD8+ T cells in the brain 56. Reichmann G, Villegas EN, Craig L, Peach R, Hunter CA (1999) The CD28/B7 interaction
and visualization of a kinesis-associated system of reticular fibers. Immunity 30: is not required for resistance to Toxoplasma gondii in the brain but contributes to the
300–311. development of immunopathology. J Immunol 163:3354–3362.
27. Barber DL, et al. (2006) Restoring function in exhausted CD8 T cells during chronic 57. Pfaffl MW (2001) A new mathematical model for relative quantification in real-time
viral infection. Nature 439:682–687. RT-PCR. Nucleic Acids Res 29:e45.

Bhadra et al. PNAS | May 31, 2011 | vol. 108 | no. 22 | 9201

You might also like