2017 - Oxidative Stress Controls Regulatory T Cell Apoptosis and Suppressor Activity and PD-L1-Blockade Resistance in Tumor

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Articles

Oxidative stress controls regulatory T cell apoptosis


and suppressor activity and PD-L1-blockade
© 2017 Nature America, Inc., part of Springer Nature. All rights reserved.

resistance in tumor
Tomasz Maj1,9, Wei Wang1,2,9, Joel Crespo1,3, Hongjuan Zhang1, Weimin Wang1,4, Shuang Wei1, Lili Zhao5,
Linda Vatan1, Irene Shao1, Wojciech Szeliga1, Costas Lyssiotis6,7, J Rebecca Liu4, Ilona Kryczek1 & Weiping Zou1,3,7,8

Live regulatory T cells (Treg cells) suppress antitumor immunity, but how Treg cells behave in the metabolically abnormal tumor
microenvironment remains unknown. Here we show that tumor Treg cells undergo apoptosis, and such apoptotic Treg cells
abolish spontaneous and PD-L1-blockade-mediated antitumor T cell immunity. Biochemical and functional analyses show that
adenosine, but not typical suppressive factors such as PD-L1, CTLA-4, TGF-b, IL-35, and IL-10, contributes to apoptotic Treg-
cell-mediated immunosuppression. Mechanistically, apoptotic Treg cells release and convert a large amount of ATP to adenosine
via CD39 and CD73, and mediate immunosuppression via the adenosine and A2A pathways. Apoptosis in Treg cells is attributed
to their weak NRF2-associated antioxidant system and high vulnerability to free oxygen species in the tumor microenvironment.
Thus, the data support a model wherein tumor Treg cells sustain and amplify their suppressor capacity through inadvertent death
via oxidative stress. This work highlights the oxidative pathway as a metabolic checkpoint that controls Treg cell behavior and
affects the efficacy of therapeutics targeting cancer checkpoints.

Extensive studies have been conducted to define the development, The homeostatic balance between Treg cells and T helper cells may
conversion, stability, and regulatory mechanisms of CD4+Foxp3+ Treg be metabolically regulated in mice20–23. However, Treg cells adopt
cells in homeostasis and a variety of disease models 1–10. It is well memory and effector phenotypes in the human tumor microenvi-
known that Treg cells are recruited into the tumor microenvironment ronment4,24. It is unknown whether Treg cells are subject to glycolytic
and act as one of the major immunosuppressors dampening sponta- regulation in tumors. Furthermore, oxidative stress is an additional
neous tumor-associated antigen (TAA)-specific T cell immunity 4–6, metabolic feature in the tumor microenvironment. Recent studies
as well as immunotherapy-induced and active-vaccination-induced have shown that myeloid dendritic cells (DCs) are phenotypically and
antitumor immunity5,6. However, how Treg cells behave in the meta- functionally altered by oxidative stress in the tumor microenviron-
bolically abnormal tumor microenvironment remains unknown. ment25. However, it is unknown whether oxidative stress alters Treg
The Warburg effect is an important metabolic feature in many types cell phenotype and function in tumors.
of cancer11. Recent studies indicate that glycolysis regulates T cell To address these questions, we examined the phenotypic and func-
activation and effector function12,13. Given that glucose, among other tional nature of Treg cells in the tumor microenvironment in human
nutrients, is poorly replenished in tumors, it is assumed that T cell ovarian cancer and in several types of mouse cancer, and investigated
glycolytic metabolism is altered as a result of the Warburg effect in the mechanisms and roles of metabolism in shaping the biological
the tumor microenvironment13–16. In support of this, poor glycolysis behaviors of Treg cells. We observed that Treg cells were highly apoptotic
can alter effector memory T cell function in the tumor microenvi- in the tumor microenvironment, and that apoptotic Treg cells achieved
ronment14,16. In addition, oxygen-sensing prolyl-hydroxylase pro- superior suppressor function via an oxidative-stress-associated
teins17, potassium ions released from necrotic cells18, and abnormal mechanism. Furthermore, we found that oxidative stress, rather than
zinc metabolism19 can impair effector T cell function in the tumor glycolysis, was the metabolic mechanism that controlled tumor Treg
microenvironment. These findings underscore the significance of the cell functional behavior and tempered the therapeutic efficacy of
metabolic regulation of memory T cells in tumors. immune checkpoint therapy.

1Department of Surgery, University of Michigan School of Medicine, Ann Arbor, Michigan, USA. 2Department of Immunology and Key Laboratory of Medical
Immunology of the Ministry of Public Health, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China. 3Rackham Graduate School,
University of Michigan, Ann Arbor, Michigan, USA. 4Department of Obstetrics and Gynecology, University of Michigan School of Medicine, Ann Arbor, Michigan,
USA. 5Department of Biostatistics, University of Michigan School of Medicine, Ann Arbor, Michigan, USA. 6Department of Molecular and Integrative Physiology,
University of Michigan School of Medicine, Ann Arbor, Michigan, USA. 7The University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor,
Michigan, USA. 8Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA. 9These authors contributed equally to this work.
Correspondence should be addressed to W.Z. (wzou@med.umich.edu).

Received 20 September; accepted 5 October; published online 30 October 2017; doi:10.1038/ni.3868

1332 VOLUME 18 NUMBER 12 DECEMBER 2017 nature immunology


Articles

RESULTS more potent suppressors than live Treg cells (Fig. 2c). Furthermore,
High Treg cell apoptosis in the tumor microenvironment mouse apoptotic tumor-infiltrating Treg cells, rather than apoptotic
A previous study showed that Treg cells are recruited into the human tumor-infiltrating conventional T cells, strongly inhibited T cell IL-2
tumor microenvironment and inhibit TAA-specific T cell immunity4. production (Fig. 2d). Thus, mouse tumor-associated apoptotic Treg
However, it is unknown how Treg cells behave in the metabolically cells are powerful immune suppressors.
abnormal tumor microenvironment. To investigate this, we used In addition to in vitro and ex vivo suppressor activity, we studied
polychromatic flow cytometry analysis (Supplementary Fig. 1a) whether apoptotic Treg cells suppress tumor immunity in vivo, using
to analyze cell proliferation and apoptosis in primary Foxp3+ Treg three systems: (i) spontaneous tumor immunity, (ii) tumor immu-
cells and conventional Foxp3−CD4+ T cells in human ovarian cancer nity induced by PD-L1-checkpoint blockade, (iii) and adoptive TAA-
tissues. We found that Treg cells expressed higher levels of the cell specific T-cell-transfusion-mediated tumor immunity. In the first
cycle protein Ki67 than conventional T cells did in the human ovar- model, we treated mice carrying MC38 cancer cells with apoptotic Treg
ian cancer microenvironment (Supplementary Fig. 1b). In addition, cells. We found that tumor volumes were larger in mice that received
flow cytometry analyses showed that Treg cells underwent substan- apoptotic Treg cells compared with those in control mice (Fig. 2e).
© 2017 Nature America, Inc., part of Springer Nature. All rights reserved.

tial apoptosis compared with Foxp3− conventional T cells in primary Then, we examined the potential effect of apoptotic Treg cells on anti-
and metastatic ovarian cancer tissues, as determined on the basis PD-L1 therapy in this model. PD-L1 blockade reduced tumor growth
of cleaved caspase-3 expression (Fig. 1). Immunofluorescence stain- (Fig. 2e) and enhanced the expression of effector T cell cytokines in
ing demonstrated the colocalization of Foxp3 and cleaved caspase-3 tumor-draining lymph nodes (Fig. 2f) and tumor tissue (Fig. 2g).
in ovarian cancer tissues (Fig. 1c and Supplementary Fig. 1c). We However, apoptotic Treg cells abolished these antitumor effects medi-
quantified proapoptotic and antiapoptotic gene transcripts in human ated by anti-PD-L1 (Fig. 2e–g). Thus, apoptotic Treg cells inhibit spon-
ovarian-cancer-infiltrating Treg cells and conventional T cells. Treg taneous and checkpoint-blockade-induced antitumor immunity.
cells expressed high levels of proapoptotic gene transcripts (Fig. 1d) Finally, we explored the role of apoptotic Treg cells in two adoptive
and low levels of antiapoptotic gene transcripts (Fig. 1e) compared TAA-specific T cell transfer models. We transfused MHC class I–
with conventional T cells in the same human ovarian cancers. We restricted (ovalbumin (OVA)-specific; OT-I) T cell receptor–transgenic
obtained similar results in mice with ID8 ovarian cancer, MC38 colon T cells into ID8-OVA-bearing (otherwise wild-type) mice and
cancer, and B16 melanoma (Fig. 1f,g). Furthermore, using gene set then treated the mice with apoptotic Treg cells and live Treg cells.
enrichment analysis, we found enriched expression of apoptotic Both apoptotic Treg cells and live Treg cells promoted tumor growth
genes in infiltrating Treg cells in mouse B16 melanoma (Fig. 1h and (Supplementary Fig. 2d) and inhibited effector T cell cytokine
Supplementary Tables 1 and 2) in the GEO database (GSE55705)26. expression (Supplementary Fig. 2e,f). However, apoptotic Treg cells
In mouse Treg cells cultured with MC38 tumor media, we detected demonstrated superior suppressor activity compared with that of live
increased expression of proapoptotic genes (Supplementary Fig. 1d) Treg cells (Supplementary Fig. 2d–f). In addition, we transferred T
and decreased expression of antiapoptotic genes (Supplementary cell receptor–transgenic CD8+ T cells that specifically recognized
Fig. 1e). Thus, the data indicate that Treg cells are highly apoptotic in the melanoma antigen PMEL into B16-F10-bearing Rag2−/− mice.
the tumor microenvironment. We treated the mice with apoptotic Treg cells and anti-PD-L1 mAb
(Supplementary Fig. 2g). Again, we found that apoptotic Treg cells
Apoptotic Treg cells mediate superior immunosuppression suppressed PMEL-specific T cell immunity and disabled the antitu-
Foxp3 controls Treg cell development and functional integrity1,2. Live mor effect of PD-L1 blockade, as shown by increased tumor growth
Treg cells inhibit TAA-specific T cell activation 4,6,27. It is unknown (Fig. 2h), shortened mouse survival (Fig. 2i), and reduced numbers
whether apoptotic Treg cells are functionally suppressive. To test of IFN-γ+TNF+ polyfunctional effector T cells (Fig. 2j,k). Thus,
this, we isolated infiltrating Treg cells from human ovarian cancer apoptotic Treg cells were superior suppressors in multiple in vitro and
samples, induced their apoptosis with anti-Fas monoclonal antibody in vivo models.
(mAb) (Supplementary Fig. 2a), and examined their suppressor
activity. Apoptotic Treg cells inhibited the expression of interferon-γ Apoptotic Treg cells make suppressive nonprotein factor(s)
(IFN-γ) and tumor necrosis factor (TNF) in CD4+ and CD8+ T cells Next, we studied the cellular and molecular mechanisms by which
(Fig. 2a). When we compared the suppressor activities of live and apoptotic Treg cells suppress T cell activation. Myeloid cells, including
apoptotic infiltrating Treg cells and apoptotic conventional T cells macrophages, can process apoptotic cells and may mediate immu-
in human ovarian cancer, we found that apoptotic Treg cells were nosuppression in the tumor microenvironment28,29. To explore
more efficient than live Treg cells at inhibiting T cell IL-2 production whether apoptotic Treg cell–mediated immunosuppression depends
(Fig. 2b). Thus, human-cancer-associated apoptotic Treg cells are on macrophages, we tested the effect of apoptotic Treg cells on tumor
potent immune suppressors. immune response with macrophage depletion in an MC38 model.
Next, we carried out similar experiments with Treg cells from As expected, the addition of apoptotic Treg cells and the depletion
healthy mice and tumor-bearing mice. As expected, in the in vitro T of macrophages promoted and inhibited tumor growth, respectively
cell suppressive assay, live Treg cells from healthy mice inhibited T cell (Fig. 3a). Accordingly, polyfunctional effector T cell cytokine
cytokine production (Supplementary Fig. 2b,c). We induced Treg cell expression was suppressed after the addition of apoptotic Treg cells
apoptosis with irradiation, serum starvation, fluorouracil, and anti- and enhanced after macrophage depletion (Fig. 3b,c). Apoptotic
Fas mAb. Similar to what we observed in human apoptotic Treg cells Treg cells remained immunosuppressive after macrophage depletion
(Fig. 2b), regardless of how Treg cell apoptosis was induced, mouse (Fig. 3a–c). These observations suggest that macrophages are dispensable
apoptotic Treg cells were superior at suppressing the expression of for apoptotic Treg cell–mediated immunosuppression in this system.
T cell TNF and IL-2 compared with identical amounts of normal live On the molecular level, live Treg cells express Foxp3 and can
Treg cells (Supplementary Fig. 2b,c). We further compared the sup- mediate immunosuppression via multiple modes of action5,30,31.
pressor activities of live and apoptotic mouse MC38-tumor-infiltrating Immunoblotting showed high levels of immunosuppression-associated
Treg cells. Again, apoptotic mouse tumor-infiltrating Treg cells were membrane molecules, including PD-L1, PD-1, CTLA-4, CD39,

nature immunology VOLUME 18 NUMBER 12 DECEMBER 2017 1333


Articles

a Foxp3+ Foxp3–
b c 55 µm g Foxp3–CD4+
Foxp3–CD4+ Foxp3+CD4+
Foxp3+CD4+ 70
23 12 DAPI Foxp3

Primary
*
30 * 60

Cleaved caspase-3+ (%)


* *
50

Annexin V+ (%)
25
20
20 µm cCASP3 CD3 *
40

Metastatic
35 4 15 30
Events

10
20
5
2 3 10
0 10 10 0
Cleaved caspase-3 PE Primary Metastatic 0
20 µm 20 µm ID8 MC38 B16.F10

d 20
* *
* * f lo lo
ID8 ascites FSC SSC CD45 CD3 CD4 CD8
+ + + –
h
© 2017 Nature America, Inc., part of Springer Nature. All rights reserved.

*
(fold increase)
Treg vs. Tconv

15 * Foxp3
+
Foxp3

Foxp3
+
Foxp3

0.7 GO term: “apoptosis induction”

Running enrichment score


2
10 * 3 9 4 24 11
0.6
5 0.5 ES = 0.401
0.4 P < 0.001
0 18 6 0.3
0.2
BB 11
S
C P8
C P3

P9

3
L2 X

C
FA
B C BA
L
AS
AS

AS

lo lo + + + –
0.1
C

MC38 intratumor FSC SSC CD45 CD3 CD4 CD8 0


e 1.00 *
* Foxp3
+
Foxp3– Foxp3
+
FoxpP3
– –0.1
–0.2
5
10 6 13 9 4
(fold increase)

0.75

00

00

00
Treg vs. Tconv

23 12

00

00

00

00
4
10

,0

,0

,0
*

2,

4,

6,

8,

10

12

14
3
0.50 10
Rank in gene list
7-AAD

2
10
0 55 21
0.25
3 4 5
–60 0 10
2 1 –1
0 10 10 10
Correlation with phenotype
0 Annexin V PE Cleaved caspase-3
M 1
L1
BC L2
L
L2

C
BC

Figure 1 Treg cells are highly apoptotic in the tumor microenvironment. (a,b) Apoptotic Foxp3+ Treg cells and Foxp3− conventional T cells in human
primary and metastatic ovarian cancers. Fresh human ovarian-cancer-infiltrating T cells were stained for viability, surface markers, Foxp3, and cleaved
caspase-3. Polychromatic flow cytometry analysis was used to analyze cleaved caspase-3 expression in live CD45 +CD3+CD4+ cells. The representative
histogram (a) and summarized data (b) show the relative amount of cleaved caspase-3+ cells. Data in b are the mean and s.e.m. for n = 5 subjects. PE,
phycoerythrin. (c) Cleaved caspase-3+Foxp3+ Treg cells in human ovarian cancer tissues. Ovarian cancer tissue sections were stained for CD3 (blue),
Foxp3 (red), cleaved caspase-3 (cCASP3; green), and nuclei (DAPI; white). The upper left image shows merged staining, and the white rectangle
outlines the region of interest with high T cell infiltration. Single fluorescence channels for each antigen and DAPI are shown in the upper right. For
detailed analysis (bottom), we overlaid the same region with the CD3 channel (blue) and the Foxp3 channel (red) (lower left) or with the CD3 channel
(blue) and the cleaved caspase-3 channel (green) (lower right). The arrows indicate the same cells in both of the bottom images. Red arrows, Foxp3 +
cells; green arrows, cleaved caspase-3+ cells; white arrows, triple-positive (Foxp3+cCASP3+CD3+) cells. (d,e) The expression of proapoptotic (d) and
antiapoptotic (e) genes in human ovarian-cancer-infiltrating Treg cells. Results are shown as the fold change in expression of each gene in Treg cells
after normalization to expression in conventional T cells (Tconv) isolated from the same cancer tissue. Data shown are the mean and s.e.m. of n = 5
individual subjects. (f,g) Apoptotic Treg cells and conventional T cells in mouse tumor tissues. We prepared single-cell suspensions from mouse ID8
ovarian cancer, MC38 colon cancer, and B16 melanoma and stained the cells for annexin V, 7-AAD, and cleaved caspase-3. The representative plots in f
show the percentages of apoptotic cells in Foxp3+CD4+ and Foxp3−CD4+ T cells as assessed on the basis of annexin V or cleaved caspase-3 expression.
Numbers in corners indicate the percentage of cells in the gate. The results shown in g are the mean percentages (+ s.e.m.) of annexin V+ cells among
all cells in the group; n = 5 mice. (h) Gene set enrichment analysis of apoptosis genes in tumor-associated Treg cells versus conventional T cells from
the GSE55705 data set. The enrichment score (ES) and P value are reported for the GO term “apoptosis induction” and were calculated by GSEA with
weighted enrichment statistics and ratio of classes for the metric as input parameters. Images and data are pooled from 5 experiments (b,d,e) or from 1
experiment with 5 animals (g) or are representative of single experiments with 5 subjects (a,f), 10 subjects (c), or 2 subjects per group (h). *P < 0.05,
Wilcoxon test (b) or Student’s t-test (g); *P < 0.01, Wilcoxon test (d,e).

and CD73, in apoptotic Treg cells compared with the amounts in expression in a dose-dependent manner (Supplementary Fig. 3d).
conventional T cells, regardless of their live status (Fig. 3d). PD-1 and However, neutralization of the immunosuppressive cytokine TGF-
PD-L1 blockade with specific antibodies had no effect on the suppres- β (Supplementary Fig. 3e–g), IL-35 subunit EBI3 (Supplementary
sive properties of apoptotic Treg cells (Fig. 3e). Apoptotic Treg cells Fig. 3h–j), and IL-10 (Supplementary Fig. 3k–m) with blocking
from PD-L1-deficient (Cd274−/−) (Fig. 3f), PD-1-deficient (Pdcd1−/−) antibodies did not rescue this suppression. Thus, apoptotic Treg cells
(Fig. 3g), and wild-type mice suppressed T cell IL-2 expression to release unknown soluble immunosuppressive factor(s), and TGF-β,
similar degrees (Fig. 3f,g). CTLA-4 blockade did not change the sup- IL-35, and IL-10 are not involved in apoptotic Treg cell–mediated T
pressive activity of apoptotic Treg cells (Supplementary Fig. 3a–c). cell suppression.
In addition to membrane proteins, it is possible that, similarly to live To identify the soluble suppressive factor(s), we divided the apop-
Treg cells, apoptotic Treg cells may release soluble factors including totic Treg cell supernatants into two fractions with a 3-kDa molecular-
cytokines to mediate T cell suppression. To test this possibility, we weight-cutoff filter. We observed that the fraction above 3 kDa did not
collected apoptotic Treg cell–derived supernatants via ultracentrifuga- suppress T cell IL-2 production, whereas the fraction below 3 kDa did
tion. The supernatants from apoptotic Treg cells inhibited T cell IL-2 (Fig. 3h). This molecular weight is consistent with the observations

1334 VOLUME 18 NUMBER 12 DECEMBER 2017 nature immunology


Articles

that PD-L1, PD-1, CTLA-4, IL-10, TGF-β, and IL-35 were not involved or trypsin (Fig. 3j). However, incubation with dextran-coated charcoal
in the suppressive function of apoptotic Treg cells (Fig. 3e–g and resulted in a loss of immunosuppression (Fig. 3k). The data suggest
Supplementary Fig. 3). Supernatants from apoptotic Treg cells remained that the soluble immunosuppressive factors may be small molecules,
immunosuppressive after treatment with proteinase K (Fig. 3i) rather than proteins or large peptides.

a b f Control + IgG Anti-PD-L1


g Control + IgG Anti-PD-L1
Control Apoptotic Treg Live Treg Apoptotic Tconv 1,200 84 20 5 6 0.5 12 2
12 28 11 16 9 20 10 24 41 57 28 39
900 *

IL-2 (pg/ml)
19 9 12 15
*

CD8
600

104 300 Apoptotic Treg Apoptotic Treg


1 9 1 5 1 4 Apoptotic Treg + anti-PD-L1 Apoptotic Treg + anti-PD-L1
IFN-γ PE-Cy7

103 1 7
66 58 55 59 104 104
© 2017 Nature America, Inc., part of Springer Nature. All rights reserved.

IFN-γ PE-Cy7
3 0.1

IFN-γ PE-Cy7
3 3 53 5 0.2

CD4
102 103 21 28 103 22 20

to reg

g
Li rol

re
101 102

op T

T
102

t
on

Ap ve

tic
100

C
101 101
100 101 102 103 104 100 100
TNF PE 100 101 102 103 104 100 101 102 103 104
TNF PE TNF PE

c 1,000 d 1,000 e 800 Apoptotic h 2,500


Control Treg * Apoptotic Treg
*

Tumor volume (mm3)


750 750 2,000 *
*
IL-2 (pg/ml)
IL-2 (pg/ml)

* Apoptotic Treg
Tumor volume (mm3)

600 Isotype * Apoptotic Treg


500 500 Apoptotic Treg 1,500 * + anti-PD-L1
* Anti-PD-L1 + anti-PD-L1 **
250 * 250 400 * Control
* *
1,000
Control
0 0
* 500 * *
m reg

200
to onv
to um rol

g
ap tic T l
to tro

*
re

re
T

Anti-PD-L1
T

Anti-PD-L1
T

*
t nt

op c
op on
tic or

or

tic
Ap Live Co

0
tu

0 0 7 14 21 28
ap
or
op

or

0 5 10 15 20 25 Time after tumor inoculation (d)


m
m
Tu
Tu

Time after occulation (d)

i 100 Anti-PD-L1 j Control + IgG


6 0.5
Anti-PD-L1
12 2
k – Apoptotic Treg
+ Apoptotic Treg
Control 28 39
80 30
Apoptotic Treg
Survival (%)

60 + anti-PD-L1
TNF+IFN-γ+ (%)

Apoptotic Treg 20 *
40 Apoptotic Treg + anti-PD-L1
104
3 0.1 5 0.2
IFN-γ PE-Cy7

20 103 22 20 10
102
*
Apoptotic Treg
0 101
0 7 14 21 28 100 0
100 101 102 103 104
Isotype Anti-PD-L1
Time after tumor inoculation (d)
TNF PE

Figure 2 Apoptotic Treg cells are immunosuppressive. (a) Immunosuppression mediated by human ovarian-cancer-associated apoptotic Treg cells.
CD3+CD4+CD25hi Treg cells were enriched and sorted from fresh ovarian cancer tissues. We used anti-Fas mAb to induce apoptosis in Treg cells and
conventional T (Tconv) cells. Apoptotic Treg cells, live Treg cells, and apoptotic Tconv cells were cultured with T cells at a ratio of 1:2 in the presence of
anti-CD3 and anti-CD28. Shown are representative results of flow cytometry analysis of the expression of IFN-γ and TNF in CD4+ and CD8+ T cell gates.
(b) Immunosuppression by human ovarian-cancer-associated live Treg cells and apoptotic Treg cells. We measured T cell IL-2 by ELISA on day 3.
(c) Immunosuppression mediated by mouse MC38-associated live and apoptotic Treg cells. Mouse GFP+Foxp3+ Treg cells were sorted from MC38
tumor tissues and treated with anti-Fas mAb to induce apoptosis. T cell immunosuppressive assays included equal numbers of live Treg cells and
apoptotic Treg cells. We measured T cell IL-2 by ELISA on day 3. (d) Immunosuppression mediated by mouse MC38-associated apoptotic Treg cells and
apoptotic conventional T cells. Mouse GFP+Foxp3+ Treg cells and GFP−Foxp3− T cells were enriched and sorted from MC38 tumor tissues and treated
with anti-Fas mAb to create apoptotic Treg cells and conventional T cells. T cell immunosuppressive assays included equal numbers of apoptotic Treg
cells and apoptotic conventional T cells. We measured T cell IL-2 by ELISA on day 3. In b,c,d, each individual data point represents a single donor
(n = 5), and significant differences are relative to the control. (e) The effect of apoptotic Treg cells on MC38 tumor growth in vivo. C57BL/6 mice were
inoculated subcutaneously with MC38 tumor cells or MC38 tumor cells plus radiation-induced apoptotic Treg cells. On days 7 and 14 the mice were
treated with anti-PD-L1 or isotype control antibody. Tumor volume was measured at the indicated time points. The images in the upper left show
representative tumors at the endpoint of the experiment. Data shown are the mean and s.d. from n = 10 mice per group. (f,g) Cytokine expression as
measured by flow cytometry in tumor-draining lymph nodes (f) and tumor tissue (g) at the endpoint of the experiments. The cells were gated on the
CD45+CD3+ population. n = 5 mice per group. (h,i) The effect of apoptotic Treg cells on B16-F10 tumor growth in a TAA-specific T cell transfusion
model. Experiments are described in Supplementary Figure 6g. Rag2−/− mice were inoculated subcutaneously with B16-F10 cells, and after 7 d PMEL-
specific T cells were transferred intravenously. Mice received intratumoral Treg cells and mAb treatment. Tumor growth (h) and mouse survival rate (i)
were monitored. The data in h are the mean ± s.d. of n = 10 mice per group. Survival curves with initial animal numbers were considered as 100% (i).
(j,k) The effect of apoptotic Treg cells on effector cytokine expression in PMEL-specific T cells. We used flow cytometry to analyze cytokine expression
in tumor-infiltrating CD45+CD3+CD8+ T cells. Data are presented as representative dot plots (h) and quantification of polyfunctional TNF+IFN-γ+
populations (i). The data in i are the mean and s.d. of n = 10 mice per group. Data are from 4 experiments (a), 1 experiment with 5 donors (b–d), or
1 experiment with 10 animals per group (e–k). In a,f,g,j, numbers in upper right corners show the percentage of cells in the indicated quadrants. PE,
phycoerythrin; Cy7, indotricarbocyanine. *P < 0.05 versus control, Wilcoxon test (b,d) or Student’s t-test (c,e,h,k).

nature immunology VOLUME 18 NUMBER 12 DECEMBER 2017 1335


Articles

Apoptotic Treg cells mediate suppression via the A2A pathway and CD73 and can convert ATP to adenosine32. Live human cancer-
We attempted to define the molecular nature of apoptotic Treg cell– infiltrating Treg cells express CD39 and CD73 and inhibit effector T
derived suppressive supernatants. Live mouse Treg cells express CD39 cells through an adenosinergic pathway33. We noticed that mouse

a b Tumor CD45+CD3+CD8+ e
– Apoptotic Treg + Apoptotic Treg
3,000 Control Anti-PD-L1 Anti-PD-1
* 18 10 0.3 4 0.7 6 0.7 5

– Clodronate
2,500 Apoptotic Treg 15 14 21

No Treg
Tumor volume (mm3)

2,000 *
Apoptotic Treg
Control or
+ clodronate
1,500 clodronate
103
liposomes 0.4 1 0.4 1.1

Apoptotic Treg
Control 104 102
0.3 1
25 13 9 9 9.4
1,000
© 2017 Nature America, Inc., part of Springer Nature. All rights reserved.

101

+ Clodronate
103
* 2

IFN-γ PE-Cy7
500 Clodronate 0
102

TNF
–2
0 101 –1 0 1 101 102 103
0 5 10 15 20 IL-2
100
Time after inoculation (d) 100 101 102 103 104
TNF PE

c d f g

nv
g

co
re
T

T
tic

tic
nv
g

co
to

to
re
T

T
op

op
ve

ve

40 – Apoptotic Treg 30 30
Ap

Ap
Li

Li

+ Apoptotic Treg
PD-L1
30 20 20
IFN-γ+TNF+ (%)

IL-2+ (%)
IL-2+ (%)
PD-1 * *
20
* *
CTLA-4 10 10 *
*
10 CD39
0 0

g
tro

g
g
g
l

re
tro

re
re
re

T
CD73

T
T
T

on


T
on

1 –/
–/
T

W
C
W
0
C

74

cd
– Clodronate + Clodronate
d2
β-actin

Pd
C

h 400 i 600 j 1,500 – Trypsin k 1,500


– Charcoal
+ Charcoal
+ Trypsin

400
*
IL-2 (pg/ml)

1,000
IL-2 (ng/ml)

1,000
IL-2 (pg/ml)

IL-2 (ng/ml)

200
* 200 500 500
*

0 0 0 0
Control Treg Control Treg
K

a
l

kD
tro

kD

kD

se

k
as
on

na

3
>3

<3

in
C

>

<
ei
te

o
ot
o

Ap

Ap
Pr

Pr

+ Proteinase K

Figure 3 Apoptotic Treg cells mediate immunosuppression via soluble factor(s). (a–c) The effect of macrophage depletion on apoptotic Treg cell–
mediated suppression. Mice bearing MC38 and apoptotic Treg cells were treated with clodronate-loaded liposomes. The plots show tumor volume (a)
and numbers of tumor-infiltrating effector T cells (b,c). Data in a and c are the mean and s.d. of n = 10 mice per group. *P < 0.05 versus the control,
Student’s t-test. (d) The expression of immunosuppression-associated molecules in live and apoptotic T cell subsets. The indicated mouse T cell
subsets were immunoblotted with antibodies to the indicated proteins. (e) The effect of PD-L1 or PD-1 blockade on apoptotic Treg cell–mediated T cell
suppression. Apoptotic Treg cells and apoptotic conventional T cells were induced by irradiation. Apoptotic Treg cells and apoptotic conventional T cells
were cultured with normal T cells at a ratio of 1:2 in the presence of anti-CD3. We measured the expression of T cell TNF and IL-2 by flow cytometry.
(f,g) Immunosuppression mediated by PD-L1−, PD-1−, and wild-type (WT) apoptotic Treg cells. Apoptotic Treg cells from Cd274−/− (f), Pdcd1−/− (g),
and wild-type mice were cultured with normal T cells at a ratio of 1:2 in the presence of anti-CD3. We measured the expression of T cell TNF and
IL-2 by flow cytometry. Data shown are the mean and s.e.m. (n = 5 mice). *P < 0.05 compared with controls, Wilcoxon test. (h) Immunosuppression
mediated by low-molecular-weight components in apoptotic Treg cell–derived supernatants. Treg cells were treated with anti-Fas mAb for 6 h. Treg cell
supernatants were passed through a 100-kDa cutoff filter and divided into two fractions with a 3-kDa cutoff filter. The two fractions were collected for
T cell immunosuppression assays. T cell IL-2 was detected on day 3 by ELISA. The data shown are the mean and s.d. (n = 5 mice). *P < 0.05 versus
the control, ANOVA with Dunett’s post-test. (i,j) Immunosuppression mediated by nonprotein elements in apoptotic Treg cell supernatants. Apoptotic Treg
cell supernatants (apo) were treated with proteinase K (i) or trypsin (j) and subsequently used for immunosuppression assays. T cell IL-2 was detected
on day 3 by ELISA. The data shown are the mean and s.d. (n = 5 mice). ANOVA with Dunett’s post-test, *P < 0.05. (k) The effect of the removal of
small lipid-like factors on immunosuppression mediated by apoptotic Treg cell supernatants. Apoptotic Treg cell supernatants were mixed with charcoal-
treated dextran to remove small lipid-like molecules and were subsequently used for immunosuppression assays. T cell IL-2 was detected on day
3 by ELISA. The data shown are the mean and s.d. (n = 5 mice). Wilcoxon test, *P < 0.05 versus the control. Data are representative of 10 (d) or 5
(e) experiments. In b and e, numbers in the upper right show the percentage of cells in the indicated quadrant.

1336 VOLUME 18 NUMBER 12 DECEMBER 2017 nature immunology


Articles

a 5,000 * b 30 c 600 d 60 apoptotic Treg cells inhibited T cell activation. The effect was abro-
4,000 *
gated by ZM241385 (Fig. 4f,g). Thus, apoptotic Treg cells mediate

TNF cells (%)


Adenosine (nM)

immunosuppression via adenosine and the A2A pathway.

TNF cells (%)


20 400 40

IL-2 (pg/ml)
3,000 *
*
2,000 Apoptotic Treg cells release ATP and metabolize it to adenosine

+
+
10 200 20
1,000 Next, we explored how apoptotic Treg cells produce a large amount
0 0 0 0 of adenosine in the absence of exogenous ATP. Gene set enrichment
0 6 Apoptotic Treg – + +
analysis demonstrated that mouse B16-melanoma-infiltrating Treg
l

l
tro

e
tro
in

in
Time (h)
os
on

on

os
ZM241385 – – +
cells showed active expression of genes associated with purine and
en
C

en
Ad

Ad
pyrimidine metabolism (GSE55705)26 (Supplementary Fig. 5a,b),
e 40 f 40 g 30
* suggesting high energy metabolism. We hypothesized that apoptotic
* *
30 30 Treg cells generate, release, and consequently metabolize to adenosine
TNF cells (%)

IL-2+ cells (%)


IL-2+ cells (%)

20
high levels of ATP via apoptotic-cell-associated CD39 and CD73, and
© 2017 Nature America, Inc., part of Springer Nature. All rights reserved.

20
20
ultimately suppress T cell activation via adenosine in the tumor. To
+

10 10
10 test this hypothesis, we induced apoptosis in mouse Treg cells and
conventional T cells by anti-Fas treatment (Fig. 5a), serum starvation
0 0 0 (Fig. 5b), or irradiation (Fig. 5c). After the induction of apoptosis,
Apoptotic Treg – + + Apoptotic Treg – + + Apoptotic Treg – + +
Treg cells, but not conventional T cells, released a large amount of
ZM241385 – – + ZM241385 – – + ZM241385 – – +
ATP (Fig. 5a,b), possibly owing to the higher ATP content in Treg
Figure 4 Apoptotic Treg cells mediate immunosuppression via adenosine. cells (Supplementary Fig. 5c). Apoptotic cells can release small
(a) Adenosine in apoptotic Treg cell supernatants. Mouse Treg cells were molecules via pannexin-1-dependent channels36. We found that
treated with anti-Fas mAb for 6 h. (b,c) The effect of exogenous adenosine
treatment with pannexin-1 inhibitor (probenecid or carbenoxolone)
on T cell TNF (b) and IL-2 (c) expression. Mouse T cells were activated
with anti-CD3 and anti-CD28 in the absence (control) or presence of
prevented apoptotic Treg cells from releasing ATP into supernatants
adenosine (5 µM). T cell cytokines were analyzed by flow cytometry (Supplementary Fig. 5d). Thus, apoptotic Treg cells release ATP via
on day 3. (d,e) The effect of the A2A inhibitor ZM241385 on mouse pannexin-1-dependent channels.
apoptotic Treg cell–mediated immunosuppression. Mouse T cells were The amount of ATP released from apoptotic Treg cells peaked
activated with anti-CD3 and anti-CD28 in the presence of apoptotic between 0.5 and 2 h and decreased to baseline at 6–8 h after apoptosis
Treg cell supernatants for 3 d. ZM241385 was added into the culture. (Fig. 5a). This suggests that ATP may be metabolized and converted
Expression of the T cell cytokines TNF (d) and IL-2 (e) was analyzed by
flow cytometry on day 3. (f,g) The effect of ZM241385 on human cancer-
into adenosine. In line with this, the measured amounts of adenosine
associated apoptotic Treg cell–mediated immunosuppression. Human inversely correlated with those of ATP and peaked at 6 h after apop-
T cells were activated with anti-CD3 and anti-CD28 in the presence of tosis in supernatants from apoptotic Treg cells (Fig. 5d). To determine
human ovarian-cancer-associated apoptotic Treg cell supernatants for 3 d. the ability of apoptotic Treg cells to generate adenosine, we incubated
ZM241385 was added into the culture. Expression of the T cell cytokines apoptotic Treg cells in medium containing exogenous ADP or ATP. We
TNF (f) and IL-2 (g) was analyzed by flow cytometry on day 3. All data detected higher amounts of adenosine in culture medium containing
are representative of 1 experiment with cells acquired from 5 different
apoptotic Treg cells than in the control samples (Fig. 5e). We collected
animals and are shown as the mean and s.d. of n = 5 mice.*P < 0.05,
paired Student’s t-test (a–c) or ANOVA with Dunett’s post-test (d–g). In the supernatants from these experiments and filtered them with a
d–g, significant differences are versus the untreated control group. 3-kDa-cutoff filter. Again, these supernatants mediated potent sup-
pression in T cell cytokines (Fig. 5f). The data suggest that apoptotic
Treg cells can convert ADP and ATP to adenosine.
apoptotic Treg cells expressed CD39 and CD73 (Fig. 3d) and released Apoptotic Treg cells express CD39 and CD73 (Fig. 3d). To directly
a nonprotein small immunosuppressive molecule (or molecules) (Fig. determine whether CD39 and CD73 are functional in apoptotic Treg
3h–k and Supplementary Fig. 3). Thus, we hypothesized that CD39 cells, we treated apoptotic Treg cells with a CD39 inhibitor, ARL67156,
and CD73 in apoptotic Treg cells remain enzymatically active and and a CD73 inhibitor, AMP-CP. As expected, ARL67156, but not
could mediate adenosine production, and that the nonprotein small AMP-CP, prevented ATP degradation (Fig. 5g). Furthermore, CD39
immunosuppressive molecule might be adenosine. To test this hypoth- and CD73 inhibitors rescued IL-2 expression that was inhibited by
esis, we induced apoptosis in mouse Treg cells with anti-Fas mAb and apoptotic Treg cells (Fig. 5h). To genetically validate our observa-
kinetically measured adenosine in the supernatants. A colorimetric tion, we tested the suppressive activity of apoptotic Treg cells from
assay detected high amounts of adenosine at 6 h in the apoptotic Treg CD73-deficient (Nt5e−/−) mice. Live Nt5e−/− and wild-type Treg cells
cell supernatants, but not in live Treg cell medium (Fig. 4a). Mass spec- contained similar amounts of intracellular ATP (Supplementary
trometry (Supplementary Fig. 4a,b) detected a kinetic increase in Fig. 5e). Nt5e−/− and wild-type apoptotic Treg cells released similar
adenosine levels in apoptotic Treg cell supernatants (Supplementary amounts of ATP into the medium (Supplementary Fig. 5f). However,
Fig. 4b,c). The amounts of adenosine also peaked at hour 6 of Treg cell Nt5e−/− apoptotic Treg cells generated less adenosine (Supplementary
apoptosis (Supplementary Fig. 4c). Then, we examined the poten- Fig. 5g) and were less immunosuppressive than wild-type apoptotic
tial suppressive role of adenosine in apoptotic Treg cell supernatants. Treg cells (Fig. 5i,j). Next, we treated MC38-tumor-bearing mice with
The addition of exogenous adenosine to mouse T cell cultures led to apoptotic Nt5e−/− T cells and apoptotic wild-type Treg cells. We found
reduced production of TNF and IL-2 (Fig. 4b,c). A2A is a receptor that tumor volumes were larger in mice that received apoptotic wild-
for adenosine in T cells34 and mediates IL-2 suppression in particu- type Treg cells than in those administered apoptotic Nt5e−/− Treg cells
lar35. We observed that the A2A inhibitor ZM241385 abolished T cell (Fig. 5k), which indicated that apoptotic Nt5e−/− Treg cells were less
effector cytokine suppression mediated by apoptotic Treg cell superna- suppressive than apoptotic Nt5e+/+ Treg cells. In addition, we tested
tants (Fig. 4d,e). We extended these mouse studies to human ovarian whether ARL67156 (Fig. 5l) and ZM241385 (Fig. 5m) affect apoptotic
cancer. Again, the supernatants of human ovarian-cancer-infiltrating Treg cell–mediated immunosuppression in MC38-tumor-bearing mice

nature immunology VOLUME 18 NUMBER 12 DECEMBER 2017 1337


Articles

a b 2,000
c d e 1,000
6,000 * IgG 1,500 4,000 IgG * *
Tconv Tconv 800
5,000 *
* Treg 1,500 * Treg

Adenosine (nM)
3,000

Adenosine (nM)
4,000 1,000 600

ATP (nM)
*

ATP (nM)
ATP (nM)

3,000 1,000 2,000


* 400
2,000 500
500 1,000 200
1,000 *
0 0 0 0 0
0 0.5 4 6 8 Tconv Treg Tconv Treg 0 0.5 4 6 8

l
P

P
tro
AD

AT
on
Time (h) Time (h)

C
f g Untreated h 1,500 i 50 j 600
© 2017 Nature America, Inc., part of Springer Nature. All rights reserved.

600
AMP-CP
480 ARL67165 40 480
2,500 1,000
*

TNF+ cells (%)


IL-2 (ng/ml)
* *
IL-2 (pg/ml)

IL-2 (pg/ml)
360 * 30 360
* 2,000
*
ATP (nM)

1,500 20 *
240 * 240
500 *
1,000
120 10 120
500

0 0 0 0 0
0 0.5 4 6 8
l
P

to reg
e

to reg

g
g
tro

AM 56

t ol
to l
tro

re
re
op tro
on
AD

AT

T
T

T
T
C

op tr
on

71

ap otic
on

P-

ap tic
Al

ap Con

tic
ap Con

tic
Time (h)
C

L6
C

op
AR

op


T
Apoptotic Treg

e –/
e –/

W
W

t5
t5

N
N
k 2,000
* Control
l 200 – Apoptotic Treg m 200 – Apoptotic Treg
* + Apoptotic Treg * + Apoptotic Treg
1,600 * *
relative to control (%)
Tumor volume (mm3)

relative to control (%)


WT 150 150
*
Tumor volume

1,200 Tumor volume


Nt5e–/–
100 100
800

50 50
400

0 0 0

ti- pe

ti- pe
ti- pe

1
Is 1

-L AR -L1

-L AR -L1

L6 6
56
L6 6
56
–/
l
T
tro

An typ An typ

-L
-L

AR 715
AR 15
W

An oty

An otyp An oty
An oty

71
71
e

PD
PD

PD + -PD

PD
on

7
o
t5

L6
L6

Is

Is
Is
C

ti

PD +

+
+

1
e

1
o

Is
Is

ti-
ti-

Figure 5 Apoptotic Treg cells release ATP and generate high levels of adenosine. (a) Kinetics of ATP release by apoptotic Treg cells. Mouse Treg cells and
conventional T cells (Tconv) were treated with anti-Fas mAb or isotype IgG. ATP in the supernatant was measured by colorimetric assay. Mean ± s.d.,
n = 6. *P < 0.05, Student’s t-test between Treg cells and conventional T cells at each time point. (b,c) ATP released by mouse apoptotic Treg cells under
serum starvation (b) or after irradiation (c) in the presence of ectonuclease inhibitor ARL67156. The ATP concentration in apoptotic Treg supernatants
was measured at 16 h. Mean and s.d., n = 5. *P < 0.05, Student’s t-test. (d) Kinetics of adenosine generation by apoptotic Treg cells. Mouse Treg cells
and conventional T cells were treated with anti-Fas mAb or isotype IgG in the presence of the adenosine deaminase inhibitor EHNA. Adenosine was
measured in the supernatant. Mean ± s.d., n = 6. *P < 0.05, Student’s t-test between Treg cells and conventional T cells at each time point.
(e,f) Adenosine generated from exogenous ADP and ATP by apoptotic Treg cells. Exogenous ADP or ATP (5 µM) was added to fresh medium with
apoptotic Treg cells for 6 h. Adenosine was measured in the culture supernatants (e). Supernatants were used for immunosuppression assays (f). Mean
and s.d., n = 5. *P < 0.05 versus control, Wilcoxon test. (g) The effect of ARL67165 or AMP-CP on ATP release by apoptotic Treg cells. Mouse Treg cells
were treated for 8 h with anti-Fas mAb with or without ARL67165 or AMP-CP. ATP was measured in the supernatants. Mean ± s.d., n = 5. *P < 0.05
versus the untreated control, Wilcoxon test. (h) The effect of ARL67165 or AMP-CP on apoptotic Treg cell–mediated immunosuppression. Mouse Treg
cells were cultured for 8 h with anti-Fas mAb with or without ARL67165 or AMP-CP. The culture supernatants were used for T cell immunosuppression.
IL-2 was measured on day 3 by ELISA. Mean and s.d., n = 8. *P < 0.05 versus control, ANOVA with Dunett’s post-test. (i,j) Immunosuppression
mediated by wild-type (WT) and Nt5e−/− apoptotic Treg cells. Apoptotic Treg cells from wild-type and Nt5e−/− mice were used for T cell
immunosuppressive assays. TNF expression was measured by flow cytometry (i), and IL-2 was detected by ELISA (j). Mean and s.d., n = 5. *P < 0.05
versus control, Wilcoxon test. (k) The effect of Nt5e−/− apoptotic Treg cells on tumor growth in vivo. MC38 tumors were inoculated subcutaneously with
wild-type or Nt5e−/− apoptotic Treg cells. The images show representative tumors at the endpoint of the experiment. Mean and s.e.m., n = 10. Student’s
t-test, *P < 0.05 versus control. (l,m) The effects of CD39 and A2A inhibitors on tumor growth. MC38-bearing mice were treated with the CD39 inhibitor
ARL67165 (5 mg/kg, 5 d per week) or the A2A inhibitor ZM241385 (1 mg/kg, 5 d per week) with anti-PD-L1 in the presence of apoptotic Treg cells.
Mean and s.e.m., n = 10. *P < 0.05 compared with isotype control in the presence of the apoptotic Treg cell group, ANOVA with Dunnett’s post hoc test.
All data in a–j were generated in single experiments; n values for those panels indicate the number of animals used for cell isolation. Data in k–m are
from 1 experiment with 10 animals per group.

1338 VOLUME 18 NUMBER 12 DECEMBER 2017 nature immunology


Articles

a 60
b 20 Ascites c g
* NAC 1.0 * Tconv Treg
* *

mRNA (fold increase)


50 *

mRNA (fold increase)


Annexin V (%)

15 NRF2
40 0.75 Ascites
+

30 NAC
10 GCLM
20 * * 0.5
* HO-1
10 5 * *
* * 0.25 β-Actin
0 + s *
+ s

N 2
ci As trol

H C

C AC

HC

AC
N 2

2 + 2O
te sc l
2O

ci A tro
s ite

0
s te

0
A

A
te ci
on

N
on
C

P9
P8

L2

L1
1
S
P3

I1
trl
trl

AX

I1
FA

L2
C
C
2O

C
AS
AS
AS

L2
B

M
C
H

C
C
C
C
As

As

B
B
Treg Tconv

d e f h
20 H2O2
1.0 * 5 1,200
mRNA (fold increase)

NAC * Tconv *

mRNA (fold increase)


* H2O2

(relative expression)
15 1,000
© 2017 Nature America, Inc., part of Springer Nature. All rights reserved.

0.75 4 Treg
NAC

ROS (MFI)
800
3

mRNA
10 0.5 600
* * * * 2 400
5
* 0.25 *
* 1 * * 200
* *
0 0 0 0
Ctrl Casp3 Casp8 Casp9 Fas Bax Bcl2l11 Ctrl Bcl2 Bcl2l1 Mcl1 Nfe2l2 Gclm Gclc Hmox1 Nqol Ggct Tconv Treg

i 60 j 60
k 60 * l 3,000
*
50 2,500 Control
*
TNF IFN-γ (%)

Tumor volume (mm )


Annexin V (%)
Annexin V (%)

3
40 * 40 40
* * 2,000
+
+
+

30
1,500 Sulforaphane
*
+

20 20 20 Sulforaphane
10 treatment stars
1,000
0 0 0
500
ra rol

e
m

lfo D l
ra EM

C e

lfo D l
ra EM

ra rol

e
tro

tro

an
an

an

an
iu

lfo ont
lfo ont
on

on
ed

ph
ph

ph

ph

C
C

C
M

0
Su
Su

Su

Su

0 5 10 15 20 25
Ascites H2O2
Time after inoculation (d)

Figure 6 Oxidative stress induces Treg cell apoptosis in the tumor environment. (a) The effect of ovarian cancer ascites on human Treg cell apoptosis.
Mouse Treg cells and conventional T cells (Tconv) were cocultured with 50% human ovarian cancer ascites or hydrogen peroxide for 24 h. Additional
cultures were treated with N-acetyl-cysteine (NAC) as a free radical scavenger. Annexin V + Treg cells and Tconv cells were analyzed by flow cytometry.
n = 5. (b,c) The effect of ovarian cancer ascites on proapoptotic and antiapoptotic gene transcripts in Treg cells. Treg cells were exposed to ascites or
ascites plus NAC after 24 h. Proapoptotic (b) and antiapoptotic (c) transcripts were quantified by real-time PCR. n = 3. (d,e) The effect of hydrogen
peroxide on proapoptotic and antiapoptotic transcripts in Treg cells. Treg cells were exposed to hydrogen peroxide or hydrogen peroxide plus NAC after
24 h. Proapoptotic (d) and antiapoptotic (e) mouse gene transcripts were quantified by real-time PCR. n = 3. (f,g) Expression of mouse NRF2 and
NRF2-associated gene transcripts (f) and proteins (g) in T cell subsets as determined by real-time PCR and immunoblotting, respectively. n = 5. (h) The
expression of intracellular ROS in Treg cells. Treg cells and conventional T cells were cultured overnight. Amounts of intracellular ROS were measured by
flow cytometry. n = 5. MFI, mean fluorescence intensity. (i) The effect of NRF2 inducers on Treg cell apoptosis in vitro. Mouse Treg cells were cultured for
24 h with 50% ascites or medium containing H2O2. NRF2 inducers diethylmaleate (DEM; 100 µM) and sulforaphane (10 µM) were added to the culture
for 24 h. Controls were treated with vehicle (DMSO). Apoptosis was measured by flow cytometry with annexin V. n = 4. (j–l) The effect of sulforaphane
on tumor immunity. MC38-bearing mice were treated with sulforaphane (25 mg/kg, 5 d per week) or vehicle (DMSO). Tumor Treg cell apoptosis (j) and
CD8+ T cell polyfunctional cytokine expression (k) were analyzed by flow cytometry. Tumor volume was monitored (l). n = 10. All data are shown as the
mean and s.d. and are from 1 experiment with 10 animals per group (i–l) or from single experiments with cells isolated from individual animal for each
data point (a–h). *P < 0.05 versus control, Wilcoxon test (b–e), paired Student’s t-test (f,g), Student’s t-test (i–l), or Mann–Whitney U-test (a,h).

with or without PD-L1 blockade. As shown by changes in tumor vol- conventional T cells (Supplementary Fig. 6a), but not in Treg cells
ume, the two inhibitors partially subverted the immunosuppressive (Supplementary Fig. 6b). Oxidative stress disrupts DC function in
effect of apoptotic Treg cells and additionally improved the therapeutic ovarian cancer25. Thus, we tested whether oxidative stress induces Treg
outcome of PD-L1 blockade (Fig. 5l,m). Together, our data suggest cell apoptosis via reactive oxygen species (ROS) in the tumor micro-
that apoptotic Treg cells release high levels of ATP and metabolize it to environment. To this end, we treated human Treg cells with ovarian
adenosine via the CD39 and CD73 axis, and suppress T cells through cancer ascites. We found that treatment with ovarian cancer ascites
interaction with adenosine and A2A receptors. and hydrogen peroxide (H2O2) induced potent apoptosis in Treg cells,
but not in conventional T cells (Fig. 6a and Supplementary Fig. 6c).
Oxidative stress induces Treg cell apoptosis in tumors Treg cell apoptosis was reversed by treatment with the ROS scaven-
Finally, we explored why Treg cells undergo apoptosis in the tumor ger N-acetyl-cysteine (Fig. 6a). Ovarian cancer ascites (Fig. 6b,c)
microenvironment. Effector T cells are affected by glycolytic metab- and H2O2 treatment (Fig. 6d,e) led to higher expression of proapop-
olism in the tumor microenvironment13,14,16. To explore whether totic genes (Fig. 6b,d) and lower expression of antiapoptotic genes
tumor Treg cell apoptosis can be attributed to poor glycolysis, we (Fig. 6c,e) in Treg cells compared with controls. Furthermore, we
treated normal Treg cells and conventional T cells with or without detected high concentrations of the major ROS component super-
glucose and the hexokinase inhibitor 2-deoxy-d-glucose. Glucose oxide in ovarian cancer ascites (Supplementary Fig. 6d). Ovarian-
restriction and 2-deoxy-d-glucose treatment induced apoptosis in cancer-infiltrating Treg cells showed high mitochondrial activity

nature immunology VOLUME 18 NUMBER 12 DECEMBER 2017 1339


Articles

compared with that in conventional T cells (Supplementary Fig. 6e) ATP into the extracellular space via pannexin-1 channels. In line with
and produced higher amounts of intracellular ROS (Supplementary our observation, mouse apoptotic thymocytes release low-molecular-
Fig. 6f). Thus, Treg cells are relatively more sensitive to oxidative stress mass compounds via pannexin-1 channels36. Thus, apoptotic Treg cells
in the tumor microenvironment than conventional T cells are, and can self-supply and release ATP, and convert it to adenosine in the
undergo potent apoptosis. local environment. We propose a novel immune-evasion mechanism:
Next, we wondered why Treg cells are more vulnerable to oxida- tumor-infiltrating Treg cells inadvertently die to mediate, sustain, and
tive stress. The cellular antioxidant system is largely regulated by amplify powerful suppression in the tumor microenvironment.
the transcription factor NRF2 (Nfe2l2) and its associated genes37,38. It has been reported that Foxp3 is a proapoptotic factor for Treg
We quantified the expression of Nfe2l2 and NRF2-associated gene cells in mouse thymus, unless some surviving factors such as γ-chain
transcripts including Gclm, Gclc, Hmox1, Nqo1, and Ggct in nor- cytokines are present44. We have explored the potential molecular
mal Treg cells and conventional T cells. The levels of the majority mechanism of Treg cell apoptosis in the tumor microenvironment.
of these antioxidant gene transcripts were lower in mouse (Fig. 6f) Several metabolic checkpoint molecules, including AMPK, mTORC1
and human (Supplementary Fig. 6g) Treg cells than in conventional (refs. 20,21), and HIF-1α (refs. 22,23), may physiologically regulate
© 2017 Nature America, Inc., part of Springer Nature. All rights reserved.

T cells. Immunoblotting showed lower amounts of NRF2, GLCM, Treg cell function and the balance between Treg cells and effector T
and heme oxygenase (HO-1) proteins in mouse (Fig. 6g) and human cells in mouse models. However, how Treg cells (particularly human
(Supplementary Fig. 6h) Treg cells than in conventional T cells. tumor Treg cells) behave in the metabolically abnormal tumor micro-
Concentrations of intracellular ROS were higher in Treg cells than environment remains unknown. Glycolysis profoundly alters effector
in conventional T cells (Fig. 6h). Next, we tested the influence of the T cell phenotype and function in the tumor microenvironment14–16.
NRF2 inducers diethylmaleate and sulforaphane on Treg cell apop- We observed that Treg cells were not as sensitive as effector memory
tosis in the presence of ovarian cancer ascites. Both NRF2 inducers T cells to glycolytic restriction. Instead, we found that Treg cells were
reduced Treg cell apoptosis compared with that in the control (Fig. 6i). vulnerable to oxidative stress in the tumor microenvironment. Our
Furthermore, we treated MC38-tumor-bearing mice with sulforaph­ data show that free oxygen species target mitochondria and induce
ane. We found that treatment with sulforaphane reduced Treg cell apoptosis in Treg cells, consequently triggering the suppressive cascade
apoptosis (Fig. 6j), increased effector T cell cytokine expression of Treg cells in the tumor microenvironment. Thus, our current study
(Fig. 6k), and inhibited tumor growth (Fig. 6l) compared with that complements recent reports on the metabolic control of memory T
observed in the control. Taken together, these data indicate that oxida- cells14–16 and DCs25 and extends to Treg cells in human and mouse
tive stress results in apoptosis of Treg cells via ROS in the tumor micro- tumor microenvironments.
environment, which in turn increases their suppressive action. In summary, apoptotic Treg cell–mediated immunosuppression
may be a major mode of action for Treg cells in cancer and serve
DISCUSSION as a potential mechanism for checkpoint-blockade resistance.
Treg cells are recruited into the tumor microenvironment. It is thought Furthermore, Treg cell apoptosis triggered by oxidative stress is a novel
that live, but not apoptotic, Treg cells mediate immune suppres- tumor immune-evasion mechanism in the tumor microenvironment,
sion1,2,4–6. In this work, we studied Treg cells in human ovarian can- and targeting tumor metabolism is a potential therapeutic strategy
cer and in several mouse tumors. We found that Treg cells underwent for cancer treatment.
potent apoptosis in human and mouse tumor microenvironments. To
our surprise, apoptotic Treg cells were more efficient than live Treg cells Methods
at suppressing T cell activation in vitro and in vivo. More important, Methods, including statements of data availability and any associated
we demonstrated that the therapeutic efficacy of PD-L1 blockade was accession codes and references, are available in the online version of
abolished by apoptotic Treg cells in tumor-bearing mouse models. The the paper.
majority of cancer patients remain unresponsive to therapies based
Note: Any Supplementary Information and Source Data files are available in the
on PD-L1 and PD-1 signaling blockade39, and we wondered whether
online version of the paper.
this clinical unresponsiveness (or therapeutic resistance) is associated
with high degrees of Treg cell apoptosis in the tumor microenviron- Acknowledgments
ment. Furthermore, the targeting of Treg cells is considered a potential This work was supported (in part) by the US National Institutes of Health (grants
CA217540, CA123088, CA099985, CA156685, CA171306, CA190176, CA193136,
therapeutic strategy for cancer treatment. However, clinical trials with
CA211016, and 5P30CA46592 to W.Z.), the Ovarian Cancer Research Fund,
Treg cell depletion have not been translated into therapeutic efficacy and the Marsha Rivkin Center for Ovarian Cancer Research (W.Z.; I.K.). We are
in people with cancer40–43. This raises the possibility that therapeutic grateful to L. Carter and X. Hu for critical discussions about the A2A pathway. We
induction of Treg cell apoptosis may sustain and/or amplify, rather thank D. Postiff, M. Vinco, R. Craig, and J. Barikdar at the Tissue and Molecular
than disable, Treg-cell-mediated immunosuppression. Pathology Core for their assistance. We thank C. Ruan and S. Bridges at the
Metabolomics Core for their support. Cd274−/− mice and Pdcd1−/− mice were
PD-L1, CTLA-4, TGF-β, IL-10, and IL-35 are well-defined immu- provided by L. Chen (Yale University, New Haven, Connecticut, USA) and
nosuppressive components for live Treg cells1–6. Our biochemical anal- T. Honjo (Kyoto University, Kyoto, Japan), respectively.
ysis and in vivo functional studies demonstrated that these molecules
are not involved in the suppressor activity of apoptotic Treg cells. Live AUTHOR CONTRIBUTIONS
T.M., Wei Wang, I.K., and W.Z. designed the experiments. T.M., I.K.,
Treg cells express the ectoenzymes CD39 and CD73 and can convert
and W.Z. wrote the paper. Wei Wang, T.M., J.C., S.W., L.V., and I.K.
extracellular ATP to adenosine, and in turn mediate immunosup- performed the in vivo tumor experiments. L.V., W.S., I.K., and J.R.L. provided
pression via adenosine and the A2A receptor signaling pathway32. and processed clinical specimens and performed immunohistochemical and
We observed that apoptotic Treg cells release a large amount of ATP pathological analysis. T.M., Wei Wang, H.Z., I.S., and Weimin Wang performed
and efficiently convert ATP into immunosuppressive adenosine via the immunological and biochemical assays. I.K., L.Z., T.M., C.L., Wei Wang and
W.Z. analyzed data.
CD39 and CD73 in vitro and in vivo. Therefore, it can be assumed
that CD39 and CD73 remain enzymatically active in apoptotic Treg COMPETING FINANCIAL INTERESTS
cells. Furthermore, apoptotic Treg cells released a large amount of The authors declare no competing financial interests.

1340 VOLUME 18 NUMBER 12 DECEMBER 2017 nature immunology


Articles

Reprints and permissions information is available online at http://www.nature.com/ 25. Cubillos-Ruiz, J.R. et al. ER stress sensor XBP1 controls anti-tumor immunity by
reprints/index.html. Publisher’s note: Springer Nature remains neutral with regard to disrupting dendritic cell homeostasis. Cell 161, 1527–1538 (2015).
jurisdictional claims in published maps and institutional affiliations. 26. Arvey, A. et al. Inflammation-induced repression of chromatin bound by the
transcription factor Foxp3 in regulatory T cells. Nat. Immunol. 15, 580–587
1. Hori, S., Nomura, T. & Sakaguchi, S. Control of regulatory T cell development by (2014).
the transcription factor Foxp3. Science 299, 1057–1061 (2003). 27. Ko, K. et al. Treatment of advanced tumors with agonistic anti-GITR mAb and its
2. Fontenot, J.D., Gavin, M.A. & Rudensky, A.Y. Foxp3 programs the development and effects on tumor-infiltrating Foxp3+CD25+CD4+ regulatory T cells. J. Exp. Med.
function of CD4+CD25+ regulatory T cells. Nat. Immunol. 4, 330–336 (2003). 202, 885–891 (2005).
3. Chen, W. et al. Conversion of peripheral CD4+CD25− naive T cells to CD4+CD25+ 28. Cui, T.X. et al. Myeloid-derived suppressor cells enhance stemness of cancer cells
regulatory T cells by TGF-β induction of transcription factor Foxp3. J. Exp. Med. by inducing microRNA101 and suppressing the corepressor CtBP2. Immunity 39,
198, 1875–1886 (2003). 611–621 (2013).
4. Curiel, T.J. et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters 29. Wan, S. et al. Tumor-associated macrophages produce interleukin 6 and signal via
immune privilege and predicts reduced survival. Nat. Med. 10, 942–949 (2004). STAT3 to promote expansion of human hepatocellular carcinoma stem cells.
5. Sakaguchi, S. Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in Gastroenterology 147, 1393–1404 (2014).
immunological tolerance to self and non-self. Nat. Immunol. 6, 345–352 (2005). 30. Shevach, E.M. CD4+CD25+ suppressor T cells: more questions than answers. Nat.
6. Zou, W. Regulatory T cells, tumour immunity and immunotherapy. Nat. Rev. Rev. Immunol. 2, 389–400 (2002).
Immunol. 6, 295–307 (2006). 31. Vignali, D.A.A., Collison, L.W. & Workman, C.J. How regulatory T cells work. Nat.
© 2017 Nature America, Inc., part of Springer Nature. All rights reserved.

7. Li, M.O., Sanjabi, S. & Flavell, R.A. Transforming growth factor-β controls Rev. Immunol. 8, 523–532 (2008).
development, homeostasis, and tolerance of T cells by regulatory T cell-dependent 32. Deaglio, S. et al. Adenosine generation catalyzed by CD39 and CD73 expressed on
and -independent mechanisms. Immunity 25, 455–471 (2006). regulatory T cells mediates immune suppression. J. Exp. Med. 204, 1257–1265
8. Liu, Y. et al. A critical function for TGF-β signaling in the development of natural (2007).
CD4+CD25+Foxp3+ regulatory T cells. Nat. Immunol. 9, 632–640 (2008). 33. Kryczek, I. et al. Phenotype, distribution, generation, and functional and clinical
9. Maruyama, T. et al. Control of the differentiation of regulatory T cells and TH17 relevance of Th17 cells in the human tumor environments. Blood 114, 1141–1149
cells by the DNA-binding inhibitor Id3. Nat. Immunol. 12, 86–95 (2011). (2009).
10. Ouyang, W. et al. Novel Foxo1-dependent transcriptional programs control Treg cell 34. Huang, S., Apasov, S., Koshiba, M. & Sitkovsky, M. Role of A2a extracellular
function. Nature 491, 554–559 (2012). adenosine receptor-mediated signaling in adenosine-mediated inhibition of T-cell
11. Vander Heiden, M.G., Cantley, L.C. & Thompson, C.B. Understanding the Warburg effect: activation and expansion. Blood 90, 1600–1610 (1997).
the metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009). 35. Erdmann, A.A. et al. Activation of Th1 and Tc1 cell adenosine A2A receptors directly
12. Gubser, P.M. et al. Rapid effector function of memory CD8+ T cells requires an inhibits IL-2 secretion in vitro and IL-2-driven expansion in vivo. Blood 105,
immediate-early glycolytic switch. Nat. Immunol. 14, 1064–1072 (2013). 4707–4714 (2005).
13. Chang, C.H. et al. Posttranscriptional control of T cell effector function by aerobic 36. Chekeni, F.B. et al. Pannexin 1 channels mediate ‘find-me’ signal release and
glycolysis. Cell 153, 1239–1251 (2013). membrane permeability during apoptosis. Nature 467, 863–867 (2010).
14. Chang, C.-H. et al. Metabolic competition in the tumor microenvironment is a driver 37. Sporn, M.B. & Liby, K.T. NRF2 and cancer: the good, the bad and the importance
of cancer progression. Cell 162, 1229–1241 (2015). of context. Nat. Rev. Cancer 12, 564–571 (2012).
15. Ho, P.-C. et al. Phosphoenolpyruvate is a metabolic checkpoint of anti-tumor T cell 38. DeBerardinis, R.J. & Chandel, N.S. Fundamentals of cancer metabolism. Sci. Adv.
responses. Cell 162, 1217–1228 (2015). 2, e1600200 (2016).
16. Zhao, E. et al. Cancer mediates effector T cell dysfunction by targeting microRNAs 39. Zou, W., Wolchok, J.D. & Chen, L. PD-L1 (B7-H1) and PD-1 pathway blockade for
and EZH2 via glycolysis restriction. Nat. Immunol. 17, 95–103 (2016). cancer therapy: mechanisms, response biomarkers, and combinations. Sci. Transl.
17. Clever, D. et al. Oxygen sensing by T cells establishes an immunologically tolerant Med. 8, 328rv4 (2016).
metastatic niche. Cell 166, 1117–1131 (2016). 40. Barnett, B., Kryczek, I., Cheng, P., Zou, W. & Curiel, T.J. Regulatory T cells in
18. Eil, R. et al. Ionic immune suppression within the tumour microenvironment limits ovarian cancer: biology and therapeutic potential. Am. J. Reprod. Immunol. 54,
T cell effector function. Nature 537, 539–543 (2016). 369–377 (2005).
19. Singer, M. et al. A distinct gene module for dysfunction uncoupled from activation 41. Dannull, J. et al. Enhancement of vaccine-mediated antitumor immunity in cancer
in tumor-infiltrating T cells. Cell 166, 1500–1511 (2016). patients after depletion of regulatory T cells. J. Clin. Invest. 115, 3623–3633
20. Delgoffe, G.M. et al. The mTOR kinase differentially regulates effector and regulatory (2005).
T cell lineage commitment. Immunity 30, 832–844 (2009). 42. Attia, P., Maker, A.V., Haworth, L.R., Rogers-Freezer, L. & Rosenberg, S.A. Inability
21. Zeng, H. et al. mTORC1 couples immune signals and metabolic programming to of a fusion protein of IL-2 and diphtheria toxin (denileukin diftitox, DAB389IL-2,
establish Treg-cell function. Nature 499, 485–490 (2013). Ontak) to eliminate regulatory T lymphocytes in patients with melanoma.
22. Shi, L.Z. et al. HIF1α-dependent glycolytic pathway orchestrates a metabolic J. Immunother. 28, 582–592 (2005).
checkpoint for the differentiation of TH17 and Treg cells. J. Exp. Med. 208, 43. Kurose, K. et al. Phase Ia study of FoxP3+ CD4 Treg depletion by infusion of a
1367–1376 (2011). humanized anti-CCR4 antibody, KW-0761, in cancer patients. Clin. Cancer Res.
23. Dang, E.V. et al. Control of TH17/Treg balance by hypoxia-inducible factor 1. Cell 21, 4327–4336 (2015).
146, 772–784 (2011). 44. Tai, X. et al. Foxp3 transcription factor is proapoptotic and lethal to developing
24. Saito, T. et al. Two FOXP3+CD4+ T cell subpopulations distinctly control the regulatory T cells unless counterbalanced by cytokine survival signals. Immunity
prognosis of colorectal cancers. Nat. Med. 22, 679–684 (2016). 38, 1116–1128 (2013).

nature immunology VOLUME 18 NUMBER 12 DECEMBER 2017 1341


ONLINE METHODS (10 ng/ml). Human T cells were isolated from healthy donor buffy coats (Carter
Ovarian cancer patients, cancer tissues, ascites, and cells. People diagnosed BloodCare) and purified directly with RosetteSep human T cell enrichment
with high-grade serous ovarian carcinomas were recruited for this study, and kits (StemCell Technologies). Treg cells from human tumors were isolated with
informed consent was obtained from all participants. The use of human sub- a commercially available Treg cell isolation kit (StemCell) and confirmed by
jects for this study was approved by the University of Michigan Institutional Foxp3 staining.
Review Board. We used clinical samples from people who had received no
prior anticancer therapies. Fresh tumor tissues and tumor ascites were proc- Induction of Treg cell apoptosis. Treg cells and conventional T cells (5 × 106
essed into single-cell suspensions for phenotype and functional studies. Tumor cells/ml) were incubated with anti-Fas mAb (10 µg/ml; clone Jo-1; eBioscience)
ascites were centrifuged, and cell-free fractions were frozen at −80 °C. for 15–30 min, washed, and suspended for further experiments. Apoptotic
Treg cells were also induced by 70-Gy irradiation and cultured serum-free for
Cell transfection. ID8 cells were transfected with plasmid pCI-neo-mOVA 24 h and with 5-fluorouracil for 24 h as indicated. For adenosine measurement,
(Addgene plasmid #25099)45. Lipofectamine 2000 (Invitrogen) was used for erythro-9-(2-hydroxy-3-nonyl) adenine (EHNA), an inhibitor of adenosine
delivery according to the vendor’s instructions. The cells were selected with deaminase, was used to prevent adenosine degradation over time.
geneticin (Invitrogen), and ovalbumin expression was confirmed by flow
© 2017 Nature America, Inc., part of Springer Nature. All rights reserved.

cytometry and used for in vivo mouse experiments. Apoptotic Treg cell–derived supernatants. Apoptotic Treg cell suspensions
were centrifuged at 500g for 5 min, 18,000g for 1 h, and 12,000g overnight.
Animals. Animal experiments were approved by the University of Michigan
Institutional Review Board. Wild-type female C57BL/6J, C57BL/6 Foxp3−GFP+ Immunosuppression assay. CD4+Foxp3− responder T cells were cocultured
(C57BL/6-Tg(Foxp3-GFP)90Pkraj/J), B6.SJL-PtprcaPepcb/BoyJ, C57BL/6- with live Treg cells or apoptotic Treg cells or with apoptotic Treg cell–derived
Tg(TcraTcrb)1100Mjb/J, B6.Cg-Thy1a/Cy Tg(TcraTcrb)8Rest/J, and Nt5e−/− supernatants in 96-well round-bottom plates at a ratio of 1:2 in the presence of
C57BL/6 mice (8–10 weeks old) were obtained from the Jackson Laboratories. anti-CD3 (2.5 µg/mL) and irradiated antigen-presenting cells. In some experi-
PD-L1-deficient (Cd274−/−) and PD-1-deficient (Pdcd1−/−) C57BL/6 mice ments, anti-PD-L1 (eBioscience), anti-PD-1 (eBioscience), anti-TGF-β (R&D),
were originally provided by Dr. Lieping Chen at Yale University and Dr. Tasuku anti-CTLA-4 (BD), anti-IL-10 (eBioscience), and anti-EBI3 (Millipore) block-
Honjo at Kyoto University, respectively. ing antibodies were added into the culture at a final concentration of 10 µg/ml.
Cells and culture supernatants were collected for cytokine quantification by
In vivo tumor experiments. For tumor growth experiments, the animals were flow cytometry analysis and ELISA.
inoculated intraperitoneally (i.p.) with 1.5 × 106 ID8 ovarian cancer cells, or
subcutaneously with 106 MC38 cells or 3 × 105 B16-F10 cells. When tumor Depletion of proteins and small molecules from Treg cell supernatants. To
cells were coinjected with apoptotic or live T cells, the ratio was 1:1. remove proteins from apoptotic or control Treg cell supernatants, we applied
For the PMEL-specific mouse model, T cell transfer was accomplished proteinase K or trypsin treatment for 1 h at 37 °C. Next, we inactivated enzymes
by a single intravenous injection of 106 PMEL-specific CD8+ T cells. Donor by boiling samples for 5 min and removed precipitates by centrifugation at
CD8+ T cells were collected from spleens and lymph nodes of B6.Cg-Thy1a/Cy 10,000g for 10 min at 4 °C. For the removal of small molecules including lipids,
Tg(TcraTcrb)8Rest/J mice and expanded with plate-bound anti-CD3 and solu- hormones, and nucleotides, media were treated with dextran-coated charcoal
ble anti-CD28 in the presence of recombinant mouse IL-2. Expanded T cells (DCC). Briefly, 0.5% (m/v) DCC suspension was mixed with the supernatants
were transferred to mice bearing B16-F10 tumors. Animals were treated as at a volume ratio of 5:1 and incubated at room temperature for 20 min. Next,
described in Supplementary Figure 2g. DCC was removed by centrifugation at 7,000g, 30 min, 4 °C. The supernatants
For the OVA-specific model, ID8-OVA-Luc tumor cells were inoculated into were collected for further experiments.
B6.SJL-PtprcaPepcb/BoyJ mice, which are congenic B6 animals that express
CD45.1 antigen. After 7 d, the mice received 106 OVA-specific CD45.2+CD8+ Small-molecule assays. For the ATP assay, pellets of Treg cells and conven-
T cells i.p. Animals were treated as described in Supplementary Figure 2d–f. tional T cells were directly suspended in 1 M perchloric acid and used for
10 min after the injection of 150 mg/kg body mass d-luciferin (Promega), the ATP assays or stored at −80 °C for further testing. Media were deproteinized
bioluminescence of luciferase-expressing tumors was assessed with the IVIS with 4 M perchloric acid for further use. A colorimetric ATP measurement
200 Spectrum imaging system (PerkinElmer). kit (Pierce) was used according to the manufacturer’s instructions. To meas-
Treatments with anti-PD-L1 or isotype control antibody (both from Bio X ure adenosine, we used adenosine deaminase (Sigma Aldrich) to transform
Cell) were done between days 7 and 20 (20 mg/kg i.p.). adenosine to xanthine in HClO4-deproteinized media, and then we measured
Macrophages were depleted with clodronate liposome (Liposoma). xanthine by colorimetric assay (BioVision).
Liposomes containing clodronate or control liposomes were injected into Adenosine was also detected by liquid chromatography coupled with tandem
mice on days 4, 6, 8, and 10 (200 µl i.p.). mass spectrometry. Cell culture medium was collected and analyzed freshly
All tumor cell lines used in the experiments were routinely tested for myco- in an LC–triple quadrupole mass spectrometer (Agilent 6490 series) with a
plasma contamination by PCR assay. Randomization of animals was not used jet stream electrospray ionization source (Agilent). Chromatography was car-
in experiments. No blinding was done. ried out on a Phenomenex Luna NH2 column (1 × 150 mm) and Agilent 1290
series liquid chromatograph. Mobile phase A was 5 mM ammonium acetate in
Cell culture and biochemical reagents. RPMI 1640 medium supplemented LC-MS-grade water, pH adjusted to 9.9 with ammonium hydroxide. Mobile
with 10% FBS (Invitrogen) was used for the majority of the experiments. For phase B was acetonitrile. 200 µl of extraction solvent was added to 200 µl of
cultures without glucose, the cells were treated with RPMI 1640 medium medium and vertexed briefly. A series of calibration standards (adenosine: 0,
without glucose (Sigma-Aldrich) and charcoal-treated FBS (Invitrogen). 0.01, 0.03, 0.1, 0.3, 1, and 5 µg/ml) were prepared along with the samples. The
Probenecid, carbenoxolone, diethylmaleate, l-sulforaphane, N-acetyl-cysteine, following transitions were used to identify and quantify adenosine: m/z 266.1
H2O2, ZM241385, AMP-CP, ARL67165, EHNA, ADP, ATP, and adenosine → m/z 134.1. Data were processed by MassHunter workstation software, ver-
were obtained from Sigma-Aldrich. Cell lines were obtained from the US sion B.06. Experiments were performed with biological replicates. Each data
National Institutes of Health or ATCC. All cell lines were routinely tested for point was acquired from cells harvested from different animals.
mycoplasma contamination.
ELISA. T cell culture supernatants were collected at the time points indicated
Isolation and culture of Treg cells and conventional T cells. Single-cell suspen- in the relevant figures. T cell effector cytokines were detected with ELISA
sions were prepared from mouse and human tissues including tumor tissues, kits (R&D).
tumor ascites, and tumor-draining lymph nodes as previously reported46–49.
Mouse Foxp3+GFP+ Treg cells were enriched and sorted from spleen and lymph Real-time PCR. Total RNA was isolated from Trizol-lysed (Invitrogen) sam-
nodes and/or were generated in the presence of IL-2 (10 ng/ml) and TGF-β ples according to the manufacturer’s instructions. A reverse-transcriptase

nature immunology doi:10.1038/ni.3868


reaction was performed with oligo-dT primers and AMV polymerase Tris-HCl, pH 6.8, 12.5% glycerol, 1% SDS, 0.01% bromophenol blue, 5% 2-mer-
(Invitrogen). Real-time PCR reactions were carried out with the Fast-SYBR captoethanol). Then they were analyzed by SDS–PAGE followed by western
kit (Applied Biosystems) and the primers listed in Supplementary Table 3. blotting with primary antibodies as follows: anti-human/mouse NRF2 (clone
1808Y; Abcam), anti-human/mouse GCLM (rabbit polyclonal; Abcam), anti-
Flow cytometry. Antibodies were from BD Biosciences, eBioscience, or mouse/human PD1 (clone 7A11B1; Thermo Fisher Scientific), anti-mouse PD-
Invitrogen. For human cells, we used anti-CD45 (clone HI30), anti-CD3 (SK7), L1 (goat polyclonal; R&D Systems), anti-mouse CTLA-4 (clone 63828; R&D
anti-CD4 (RPA-T4), anti-cCASP3 (9H19L2), anti-Foxp3 (PCH101), anti-TNF Systems), anti-mouse CD39 (clone 495826; R&D Systems), and anti-mouse
(MAb11), anti-IL-2 (MQ1-17H12), and anti-IFN-γ (4S.B3). Mouse cells were CD73 (clone sc-32299; Santa Cruz Biotechnology). Proper secondary HRP-
stained with anti-CD45 (30-F11), anti-CD45.1 (A20), anti-CD45.2 (104), anti- conjugated anti-rat, anti-rabbit (both from Vector Laboratories), or anti-goat
CD3 (500A2), anti-CD4 (RM4-5), anti-CD8 (53-6.7), anti-cCASP3 (9H19L2), (Santa Cruz Biotechnology) antibodies were used as secondary reagents.
anti-Foxp3 (FJK-16s), anti-TNF (MP6-XT22), anti-IL-2 (JES6-5H4), and anti-
IFN-γ (XMG1.2). For surface staining, the cells were incubated with anti- Bioinformatics. Publicly available microarray data were analyzed with GSEA
bodies for 20 min, washed, and fixed in Fix/Perm solution (BD Biosciences). software v. 2.0, which is freely available from the Broad Institute (http://www.
After being washed with Perm/Wash buffer (BD Biosciences), the cells were broad.mit.edu/gsea/)50.
© 2017 Nature America, Inc., part of Springer Nature. All rights reserved.

stained intracellularly for 30 min, washed, and fixed in 4% formaldehyde


(Sigma Aldrich). In the case of apoptosis detection in cell culture, the cells Statistics. Statistical analysis was done with R v. 3.0.2. For comparison of two
were stained with human recombinant annexin V (BD Biosciences) followed groups, we used two-sided Student’s t-test or Mann–Whitney U-test, depend-
by 7-ADD. In the case of tissue samples, the cells were stained for viability ing on the data distribution (Shapiro–Wilk test) and homogeneity of variance
with blue fluorescent fixable dye (Life Technologies) followed by annexin V (Fisher’s test or Levene’s test). Parametrical or nonparametrical ANOVA tests
and Foxp3. All samples were read on an LSR II cytometer (BD) and analyzed accompanied by two-sided post hoc analysis were used for multiple-compari-
with FACS DIVA software v. 8.0 (BD Biosciences). son experiments.

Immunofluorescence staining. Tumor tissues were fixed in 10% formalin Data availability. A Life Sciences Reporting Summary for this paper is available.
and embedded in paraffin for immunofluorescence staining. Multiplexed
fluorescence staining was done with the OpalTM 4-plex staining system
(PerkinElmer). Tissue sections were stained with anti-CD3 (clone PS1,
mouse IgG2a; Novocastra/Leica), anti–cleaved caspase-3 (rabbit polyclonal
IgG; Cell Signaling), and anti-Foxp3 (clone 259D, mouse IgG1; BioLegend). 45. Yang, J. et al. Kupfer-type immunological synapse characteristics do not predict
anti-brain tumor cytolytic T-cell function in vivo. Proc. Natl. Acad. Sci. USA 107,
The secondary antibodies used were goat anti-mouse IgG2-α conjugated with 4716–4721 (2010).
Alexa Fluor 488 or goat anti-rabbit IgG (H+L) conjugated with Alexa Fluor 46. Wang, W. et al. Effector T cells abrogate stroma-mediated chemoresistance in
568. Staining slides were analyzed with the Mantra Quantitative Pathology ovarian cancer. Cell 165, 1092–1105 (2016).
Workstation (PerkinElmer). 47. Peng, D. et al. Epigenetic silencing of TH1-type chemokines shapes tumour
immunity and immunotherapy. Nature 527, 249–253 (2015).
48. Curiel, T.J. et al. Blockade of B7-H1 improves myeloid dendritic cell–mediated
Immunoblotting. After centrifugation cells were dissolved in cell lysis buffer antitumor immunity. Nat. Med. 9, 562–567 (2003).
(50 mm Tris-HCl, pH 7.4, 1% Triton X-100, 0.2% sodium deoxycholate, 0.2% 49. Zou, W. et al. Stromal-derived factor-1 in human tumors recruits and alters the
SDS, 1 mM sodium EDTA) supplemented with protease inhibitors (5 µg/ml function of plasmacytoid precursor dendritic cells. Nat. Med. 7, 1339–1346
(2001).
leupeptin, 5 µg/ml aprotinin, and 1 mM phenylmethylsulfonyl fluoride) and 50. Subramanian, A. et al. Gene Set Enrichment Analysis: a knowledge-based approach
protein concentrations were determined with Bio-Rad protein assay rea- for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102,
gent. The lysates were boiled for 5 min in Laemmli sample buffer (50 mm 15545–15550 (2005).

doi:10.1038/ni.3868
 nature immunology
nature research | life sciences reporting summary
Corresponding author(s): Weiping Zou
Initial submission Revised version Final submission

Life Sciences Reporting Summary


Nature Research wishes to improve the reproducibility of the work that we publish. This form is intended for publication with all accepted life
science papers and provides structure for consistency and transparency in reporting. Every life science submission will use this form; some list
items might not apply to an individual manuscript, but all fields must be completed for clarity.
For further information on the points included in this form, see Reporting Life Sciences Research. For further information on Nature Research
policies, including our data availability policy, see Authors & Referees and the Editorial Policy Checklist.

` Experimental design
© 2017 Nature America, Inc., part of Springer Nature. All rights reserved.

1. Sample size
Describe how sample size was determined. No statistical method was used to calculate sample size.

2. Data exclusions
Describe any data exclusions. Sample size was estimated on the basis of similar research reported in the
literature.
3. Replication
Describe whether the experimental findings were All attempts at replication were successful.
reliably reproduced.
4. Randomization
Describe how samples/organisms/participants were The majority of the animal experiment were not randomized. Specific randomized
allocated into experimental groups. experiment was stated in figure legends.
5. Blinding
Describe whether the investigators were blinded to Blinding was not performed.
group allocation during data collection and/or analysis.
Note: all studies involving animals and/or human research participants must disclose whether blinding and randomization were used.

6. Statistical parameters
For all figures and tables that use statistical methods, confirm that the following items are present in relevant figure legends (or in the
Methods section if additional space is needed).

n/a Confirmed

The exact sample size (n) for each experimental group/condition, given as a discrete number and unit of measurement (animals, litters, cultures, etc.)
A description of how samples were collected, noting whether measurements were taken from distinct samples or whether the same
sample was measured repeatedly
A statement indicating how many times each experiment was replicated
The statistical test(s) used and whether they are one- or two-sided (note: only common tests should be described solely by name; more
complex techniques should be described in the Methods section)
A description of any assumptions or corrections, such as an adjustment for multiple comparisons
The test results (e.g. P values) given as exact values whenever possible and with confidence intervals noted
A clear description of statistics including central tendency (e.g. median, mean) and variation (e.g. standard deviation, interquartile range)
Clearly defined error bars
June 2017

See the web collection on statistics for biologists for further resources and guidance.

1
Nature Immunology: doi:10.1038/ni.3868
` Software

nature research | life sciences reporting summary


Policy information about availability of computer code
7. Software
Describe the software used to analyze the data in this R version 3.0.2 was used for statistical analysis.
study.
For manuscripts utilizing custom algorithms or software that are central to the paper but not yet described in the published literature, software must be made
available to editors and reviewers upon request. We strongly encourage code deposition in a community repository (e.g. GitHub). Nature Methods guidance for
providing algorithms and software for publication provides further information on this topic.

` Materials and reagents


Policy information about availability of materials
8. Materials availability
Indicate whether there are restrictions on availability of No unique materials were used.
© 2017 Nature America, Inc., part of Springer Nature. All rights reserved.

unique materials or if these materials are only available


for distribution by a for-profit company.
9. Antibodies
Describe the antibodies used and how they were validated Antibodies were used for flow cytometry, ELISA assays, and immunoblotting. All
for use in the system under study (i.e. assay and species). antibodies and ELISA kits were purchased from well recognized vendors and were
reported by other researchers.

10. Eukaryotic cell lines


a. State the source of each eukaryotic cell line used. The sources were stated or cited.

b. Describe the method of cell line authentication used. Cell lines (except ID8, original source was cited) were authenticated by the
providers.

c. Report whether the cell lines were tested for All cell lines in our laboratory are routinely tested for mycoplasma contamination.
mycoplasma contamination.

d. If any of the cell lines used are listed in the database No cell line used in the paper is listed in ICLAC database.
of commonly misidentified cell lines maintained by
ICLAC, provide a scientific rationale for their use.

` Animals and human research participants


Policy information about studies involving animals; when reporting animal research, follow the ARRIVE guidelines
11. Description of research animals
Provide details on animals and/or animal-derived In the study we used female mice, 6-8 weeks old. Strains used in the in vivo
materials used in the study. settings were wild-type B6, CD45.1 B6 or Foxp3-GFP mice. Cells for some
experiments were also obtained from OT-I or Pmel mice, as indicated in the
methods section and figure legends of appropriate experiments.

Policy information about studies involving human research participants


12. Description of human research participants
Describe the covariate-relevant population Fresh human tumor tissues were used for our studies. We do not link our in vitro
characteristics of the human research participants. experiments and histological staining to patient clinical characteristics. Thus, we do
not provide the population characteristics.
June 2017

2
Nature Immunology: doi:10.1038/ni.3868
nature research | flow cytometry reporting summary
Corresponding author(s): Weiping Zou
Initial submission Revised version Final submission

Flow Cytometry Reporting Summary


Form fields will expand as needed. Please do not leave fields blank.

` Data presentation
For all flow cytometry data, confirm that:
1. The axis labels state the marker and fluorochrome used (e.g. CD4-FITC).
2. The axis scales are clearly visible. Include numbers along axes only for bottom left plot of group (a 'group' is an analysis of
© 2017 Nature America, Inc., part of Springer Nature. All rights reserved.

identical markers).
3. All plots are contour plots with outliers or pseudocolor plots.
4. A numerical value for number of cells or percentage (with statistics) is provided.

` Methodological details
5. Describe the sample preparation. For all fresh cells obtained from human or animal tissues we performed
mechanical disruption of the cells (cutting with blade and pushing the
fragments of tissue through the strainer). After washing, cell suspension
was transferred to fresh tubes through the 40 um mesh to remove
remaining large fragments or clumped dead cells. Next, cells were
centrifuged in Ficol density gradient and used for cells culture or flow
cytometry staining.
When the cells were stained for surface antigens, the staining was
performed in PBS + 2 mM EDTA + 0.5% BSA. Cells were read directly or
after fixation with 2% formaldehyde in PBS. For intracellular staining, we
used commercially available reagents (BD). If needed, the cells were
stimulated with PMA/ionomycin/brefeldin cocktail for 4 hours.

6. Identify the instrument used for data collection. LSR II (BD)

7. Describe the software used to collect and analyze BD FACSDiva, version 8.0
the flow cytometry data.

8. Describe the abundance of the relevant cell When cells were sorted or enriched, the purity was confirmed by flow
populations within post-sort fractions. cytometry and in each case was above 90% purity.

9. Describe the gating strategy used. The cells were gated on FSC-A/SSC-A basis on the location known to
contain lymphoid cells. Next, CD45+ positive gating was used to exclude
tumor cells. Next, CD3+CD8+ or CD3+CD4+ cells were analyzed in CD45+
gate.
Tick this box to confirm that a figure exemplifying the gating strategy is provided in the Supplementary Information.
June 2017

1
Nature Immunology: doi:10.1038/ni.3868

You might also like