Article
CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer Graphical Abstract
Authors CX3CR1
CD4 T cell
Cytolytic CD8 T cell subset with augmented capacity to kill cancerous or virus-infected cells
T-bet Zeb2 Ly108
IL-21
+++ antigen & insufficient CD4 help
PD-1 Progenitor CD8 T cell subset with unequivocal self-renewal potential
Eomes Nr4a2
Exhausted CD8 T cell subset with limited recall potential and diminshed capacity to produce pro-inflammatory cytokines and lyse target cells
Highlights d
d
d
d
Correspondence
[email protected]
Tumor or virus-infected cell
Tcf1 Id3
Ryan Zander, David Schauder, Gang Xin, Christine Nguyen, Xiaopeng Wu, Allan Zajac, Weiguo Cui
scRNA-seq unveils a unique subset of cytolytic CD8+ T cells during chronic infection CX3CR1+ CD8+ T cells are required to control chronic viral infection CD4+ T cell help via IL-21 production is critical for CX3CR1+ CD8+ T cell formation IL-21-producing CD4+ T cells enhance CX3CR1+ TIL formation and tumor control
Zander et al., 2020, Immunity 53, 1–15 December 15, 2020 ª 2019 Elsevier Inc. https://doi.org/10.1016/j.immuni.2019.10.009
In Brief The mechanisms by which CD4+ T cell help sustains exhausted CD8+ T cells during chronic infection have remained elusive. Here, Zander and colleagues show that CD4+ T-cell-derived IL-21 is required for the formation of a distinct subset of cytolytic CX3CR1+CD8+ T cells that protect against chronic infection and cancer.
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
Immunity
Article CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer Ryan Zander,1,4 David Schauder,2,4 Gang Xin,1 Christine Nguyen,2 Xiaopeng Wu,1 Allan Zajac,3 and Weiguo Cui1,2,5,* 1Blood
Research Institute, BloodCenter of Wisconsin, Milwaukee, WI 53213, USA of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA 3Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA 4These authors contributed equally 5Lead Contact *Correspondence:
[email protected] https://doi.org/10.1016/j.immuni.2019.10.009 2Department
SUMMARY
Although CD4+ T cell ‘‘help’’ is crucial to sustain antiviral immunity, the mechanisms by which CD4+ T cells regulate CD8+ T cell differentiation during chronic infection remain elusive. Here, using singlecell RNA sequencing, we show that CD8+ T cells responding to chronic infection were more heterogeneous than previously appreciated. Importantly, our findings uncovered the formation of a CX3CR1-expressing CD8+ T cell subset that exhibited potent cytolytic function and was required for viral control. Notably, our data further demonstrate that formation of this cytotoxic subset was critically dependent on CD4+ T cell help via interleukin-21 (IL-21) and that exploitation of this developmental pathway could be used therapeutically to enhance the killer function of CD8+ T cells infiltrated into the tumor. These findings uncover additional molecular mechanisms of how ‘‘CD4+ T cell help’’ regulates CD8+ T cell differentiation during persistent infection and have implications toward optimizing the generation of protective CD8+ T cells in immunotherapy.
INTRODUCTION During chronic viral infection and cancer, CD8+ T cells undergo a differentiation process commonly referred to as T cell exhaustion (Wherry and Kurachi, 2015). This process is traditionally defined by a stepwise loss of effector functions, eventually leading to cell death (Wherry et al., 2003; Kahan et al., 2015). Despite their inability to completely clear the infection, exhausted T cells remain indispensable for viral control (Schmitz et al., 1999; Jin et al., 1999), indicating that some CD8+ T cells retain their cytotoxic potential. CD4+ T cell help has long been known to be essential for sustaining CD8+ T cell function during chronic viral infection (Battegay et al., 1994; Matloubian et al., 1994; Zajac et al., 1998). More recently, several studies have identified a critical role for CD4+ T-cell-produced interleukin-21 (IL-21)
in mediating this protective response (Elsaesser et al., 2009; Fro¨hlich et al., 2009; Xin et al., 2015). However, the underlying mechanism(s) by which CD4+ T cell-derived IL-21 coordinates this antiviral CD8+ T cell program remains incompletely understood. Previous research indicates that CD8+ T cells responding to persistent infection can exist in two distinct transcriptional states, with differential expression of the transcription factors T-bet and Eomes coordinating this cell-fate decision process (Paley et al., 2012). In addition, several recent studies identify that virus-specific CD8+ T cells are non-homogeneous and can be compartmentalized into at least two major subsets, with a CXCR5hiTCF-1hi subset serving as a self-renewing progenitor population that can give rise to a more terminally exhausted CXCR5loTCF-1lo subset (Leong et al., 2016; He et al., 2016; Im et al., 2016; Utzschneider et al., 2016). Moreover, CXCR5hiTCF-1hi CD8+ T cells are implicated as being the major responders to PD-1 checkpoint blockade (He et al., 2016; Im et al., 2016; Siddiqui et al., 2019; Kurtulus et al., 2019), a breakthrough immunotherapy demonstrated to enhance CD8+ T cell responses during chronic infections and cancer (Barber et al., 2006; Wherry and Kurachi, 2015). These observations prompted us to ask whether additional heterogeneity exists among CD8+ T cells responding to persistent viral infection and whether CD4+ T cells could regulate this multifaceted process of CD8+ T cell differentiation to better meet the needs of a chronic infection. Single-cell RNA sequencing (scRNA-seq) has emerged as a powerful technique to explore cellular diversity and differentiation states (Papalexi and Satija, 2018; Villani et al., 2017). Thus, we sought to apply scRNA-seq to this biological process in order to assess the transcriptional profiles and developmental pathways of CD8+ T cells during persistent viral infection and to determine how CD4+ T cell help regulates this intricate process of differentiation. RESULTS scRNA-Seq Reveals Broad Transcriptional Heterogeneity among LCMV-Specific CD8+ T Cells To fully characterize the heterogeneity of CD8+ T cells responding to chronic viral infection, we performed scRNA-seq on CD8+ Immunity 53, 1–15, December 15, 2020 ª 2019 Elsevier Inc. 1
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
A
B
Day 8 p.i.
Naive Slamf6+ Slamf6+Pdcd1+ D30 Pdcd1+ D30 Pdcd1+Cx3cr1+ D30 Cx3cr1+ D8 Effector D8 Effector (S phase) D8 Effector (G2/M phase)
C
Dayy 30 p.i. p
Figure 1. scRNA-Seq Reveals Transcriptional Heterogeneity among CD8 T Cells Responding to Persistent Viral Infection (A) t-SNE plot displaying clusters identified in a mixture of GP33+ CD8 T cells from day 8 and day 30 p.i. with LCMV Cl13. (B) t-SNE plots showing CD8 clusters from day 8 or day 30 time points. (C) Summary dot plot depicting relative frequency of day 8 and day 30 virus-specific CD8 T cell populations. (D) Heatmap showing Z scores for the average expression of given genes within a particular cluster. See also Figures S1 and S2.
D Day 8-specific
C3
-S
C1
C2
-N ai - S ve la la m m f6 f6 + +/ C5 C Pd -D 4cd D 30 1+ 30 Pd Pd cd cd 1+ 1+ C6 /C x3 -D cr 30 1+ Cx C7 3c r1 -D C8 + 8 Ef -D f C9 e 8 ct E -D or ffe 8 ct Ef or fe (S ct ) or (G 2/ M )
Day 30-specific
T cells specific for the GP33–41 peptide of lymphocytic choriomeningitis virus (LCMV) at days 8 and 30 post-infection (Figures 1A, S1A, and S1B). Notably, virus-specific T cells grouped distinctly into nine clusters when visualized by t-distributed stochastic neighbor-embedding analysis (t-SNE) (Figure 1A; Satija et al., 2015; Waltman and Jan van Eck, 2013), which was performed using the top nine principal components (Figures S1C and S1D). Each cluster appeared to be predominantly specific for either day 8 or day 30, with the exception of Naive (cluster 1) and Slamf6+ (cluster 2) cells, which were present at both time points in either relatively low or high frequencies, respectively (Figures 1A–1C). In addition, our analyses identified the presence of three other transcriptionally distinct clusters on day 8 postinfection (p.i.) (clusters 7–9), two of which could be distinguished by their active cell cycle phase, with clusters 8 and 9 expressing S phase and G2/M phase markers, respectively (Figures 1A–1C, S2A, and S2B). Moreover, our analyses also identified the exis2 Immunity 53, 1–15, December 15, 2020
tence of three major subsets of CD8+ T cells on day 30 p.i.: Slamf6+ cells (cluster 2); Pdcd1+ cells (cluster 4); and Cx3cr1+ cells (cluster 6) (Figures 1A–1C). Of note, the Slamf6+ subset, which was present on both day 8 and day 30, shared similarities with the previously described self-renewing progenitor population of CD8+ T cells, particularly in its expression of Tcf7, Icos, Il7r (encoding IL-7Ra), and Id3 (Im et al., 2016; Leong et al., 2016; Figures 1D, S2C, and S2D). Additionally, expression of Batf, a transcription factor we previously showed to be a critical transducer of IL-21 signaling in CD8+ T cells, was increased in this group (Figures S2C and S2D; Xin et al., 2015). Expression of Cxcr5 was also enriched within this cluster, although Tcf7 expression seemed to correlate more strongly with Slamf6 expression (Figures S2C and S2D), possibly indicating that Slamf6 might mark a larger progenitor population that contains the previously identified CXCR5+TCF-1+ subset (Utzschneider et al., 2016). Together, the Pdcd1+ and + Cx3cr1 clusters comprise what was previously thought to be a homogeneous group of terminally exhausted cells. Both of these subsets express high amounts of Gzma and Gzmb, supporting their status as functional cytotoxic cells (Figure S2G). They differed, however, in their expression of numerous inhibitory receptors, with the Pdcd1+ group expressing increased amounts of Cd244 (encoding 2B4), Cd160, Lag3, Havcr2 (encoding Tim3), and of course Pdcd1 (encoding PD-1; Figures 1D and S2E). The Pdcd1+ group could also be distinguished by their expression of Cxcr6 and Cd7, as well as the transcription factors Nr4a2, Maf, and Eomes (Figures 1D and S2E). Conversely, Cx3cr1+ cells displayed high expression of many killer cell lectin-like receptors (Klre1 and Klra9, Figures 1D and S2F; Klrd1 and Klrg1, data not shown). The Cx3cr1+ subset was also marked by high expression of the transcription factors Tbx21, Zeb2, Klf2 (Figures 1D and S2F), Id2, and Runx1 (data not shown). Overall, the transcriptional profile of Cx3cr1+ cells Ccr7 Lef1 Sell Il7r Tcf7 Id3 Slamf6 Batf Cxcr6 Nr4a2 Cd7 Cd244 Cd160 Lag3 Pdcd1 Maf Cx3cr1 Klre1 Klra9 Zeb2 Tbx21 Klrg1 Il18rap Il18r1 Ly6c2 Lgals3 Itgax Rad51 Mcm5 Cdc6 Ccna2 Ccnb2 Birc5 Cenpa
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
3
3
10
3
0
-10
-10
3
0
10
3
10
4
10
3
38.5
5
-10
3
10.8 0
10
CD44 6
10
5
10
4
10
3
10
2
10
1
10 45
0
% subset of GP33+ CD8+ T cells (%)
Liver 0.52
4.0
30
E
Bone Marrow 0.92
3
0
24.7 -10
3
5.25 0
10
3
10
4
10
36.8
14.7
43.2
58.7
2.14
2.24
81.9
13.2
5
60 40 20 0
20
60
40
20
0
80
60
40
20
0 0
10
3
10
4
0
5
10
3
-10
0
KLRG1
10
3
10
4
10
5
60
40
20
60
40
3
10
4
10
20
40
20
3
10
4
10
5
-10
-10
3
0
10
3
10
3
0
4
10
3
10
4
10
5
CD127
60
40
20
5
10
100
80
60
40
3
0
10
3
10
4
10
le
*** **
**
5
-10
3
0
10
3
10
4
10
R
PD
XC C
BATF
40
20
80
60
40
20
0 -10
3
0
10
3
10
4
10
80
60
40
20
10
10
-10
3
0
10
3
10
4
10
4
60
40
10
3
0
20
3
0
0
5
5
80
-10
0
I
100
Normalized To Mode
60
100
Normalized To Mode
80
T-bet
100
Normalized To Mode
Normalized To Mode
100
CX3CR1+ Ly108+ CX3CR1- Ly108-
T-bet
Eomes
5
-10
3
0
10
3
10
4
10
5
-10
3
0
10
3
10
4
10
** *** *** ***
0
5
5
-10
3
0
10
3
10
4
10
5
Eomes
gMFI Transcription factor
H TCF1
*** *** *** **
*** ** *** * *
400
0
-10
CX3CR1+ Ly108+ CX3CR1- Ly108-
20
0
5
*** ***
5000 4000 3000 2000 1000 800
6
10
10
80
0
0
60
CXCR5
80
0 3
0
100
Normalized To Mode
Normalized To Mode
80
-10
3
Klra9 100
100
G
80
0 -10
Normalized To Mode
3
-10
2B4 100
Normalized To Mode
40
Normalized To Mode
Normalized To Mode
Normalized To Mode
60
Normalized To Mode
Tim3 100
80
-1
PD-1 100
80
gMFI surface molecules
CXCR6 100
F
Sp
PB
L
Ly108
80
1
3
CX3CR1+ Ly108+ CX3CR1- Ly108-
100
G
-10
% CX3CR1- Ly108-
Kl ra 9 C XC R 5 C D 12 7
10
% Ly108+
R
38.6
0.66
Day
% CX3CR1+
KL
54.0
5
3
1.78
15
10
2B 4
Lung
46.7
0.35
0
4
Ti m
iLN
70
20
4
10
CX3CR1
PBL 5
10
40
GP33+ CD8+ T cell subset tissue distribution (%)
D
60
0
10
Serum viral load (PFU/ml)
10
80
Slamf6+ cells
3
Ly108
C Day 30 Pdcd1+ cells
+
CX3CR1 + Ly108 CX3CR1-Ly108-
BM
-10
1.19
4
10
0
49.5
5
10
4
N
10
Day 30 Cx3cr1+ cells
4.56
5
iL
Db-GP33
10
Li ve r Lu ng
10
en
B
CX3CR1
A
******
30000
CX3CR1+ Ly108+ CX3CR1- Ly108-
20000 10000 6000
**
*** ***
3000
J
-10
3
0
10
Ly108
9
3
10
4
10
3
0
-10
3
95.4
5
-10
3
Ly108
10
3
gMFI Granzyme B
15000
GzmB 100
80
60
40
10000
** *** **
5000
20
10
3
10
4
10
5
0
C X 3C R L 1+ R y10 1- 8+ Ly 10 8-
0
X 3C
3
4
10
86.3
1.31
96.3
0.57
t
m es Eo
Tbe 6 4 2
5
L
0
0
* * ** **
500 400
*
**
** ***
CX3CR1+ DN Ly108 CX3CR1+
300
EL4-GP33 peptide EL4-SIINFEKL (OT-1) peptide
200 100 0 0
4
8
12
Time after incubation (hours)
C
0 -10
10
IFN-γ
K Normalized To Mode
0.37 0
5
*** ***
8
Ly X 10 3C 8+ R 1Ly 10 8-
CX3CR1Ly108- 21
4
X 3C
3
-10
1.28
C
0
1.81
1+
10
7.00
R
10
3
5.43
%TNF-α+IFN-γ+
4
2.77
C
10
CD107a
CX3CR1
10
1.46
X 3C
10
CX3CR1
5
* ** ***
10
C
2
Gated on CX3CR1-Ly108-
R 1+ C Ly X 10 3C 8 + R 1Ly 10 8-
68
Gated on Ly108+
Annexin V Object Count/Wells
10
5
Gated on CX3CR1+
% CD107a+IFN-γ+
Gated on CD8+ CD44hi
BA TF
TC
F1
0
(legend on next page)
Immunity 53, 1–15, December 15, 2020 3
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
shares many similarities with short-lived effector cells found during acute infections (Dominguez et al., 2015; Kaech and Cui, 2012; Omilusik et al., 2015). We next determined how the largest subset of effector CD8+ T cells on day 8 p.i. (cluster 7; D8 effector) compared to the two major subsets of day 30 effector cells (Pdcd1+ and Cx3cr1+ clusters). To do this, we utilized two distinct computational approaches. First, we visualized the top 30 markers of each of the three clusters relative to the other two (Figure S3B). Second, a score was given to each cell based on its overall expression of the top 100 markers from each group (Figure S3C). Notably, both sets of analyses demonstrated a high degree of transcriptional divergence between the three-effector clusters. Our analyses did however identify that day 8 effector cells did resemble each of the two day 30 subsets in their expression of a few key genes (Figures S3D and S3E). Expression of the chemokine receptor Cxcr6, transcription factor Nfatc1, and inhibitory receptors Pdcd1, Cd160, and Lag3 were all shared between day 8 effector cells and the day 30 Pdcd1+ cluster (Figures S3D and S3E). Day 8 effector cells also expressed several markers of day 30 Cx3cr1+ effector cells, such as Klrc1, Itgb1, and Cx3cr1 (Figures S3D and S3E and not depicted). Therefore, though transcriptionally distinct from day 30 Pdcd1+ and Cx3cr1+ effector cells, day 8 effector cells have begun to acquire features of both of these subsets. Differential Expression of CX3CR1 and Ly108 Can Be Used to Distinguish 3 Major Subsets of CD8+ T Cells during Chronic Viral Infection We next tested whether we could use the surface markers identified from our scRNA-seq analyses to separate the three predominant populations of virus-specific CD8+ T cells that form during chronic viral infection (Figure 2A). To do this, we infected mice with LCMV Cl13 and compared expression of CX3CR1 (a cluster 6 marker) and Ly108 (encoded by Slamf6, a cluster 2 marker) on virus-specific CD8+ T cells. Using these markers, three clearly distinct subsets of CD8+ T cells emerged: Ly108+; CX3CR1Ly108; and CX3CR1+, representing clusters 2, 4, and 6 from our scRNA-seq analysis, respectively (Figures 2A, 2B, and S4A). In the spleen, approximately half of GP33-41-specific CD8+ T cells belonged to the CX3CR1+ subset, and this frequency remained relatively stable from day 8 to day 45 p.i. (Figures 2B, 2C, and S4A). On the other hand, Ly108+ and CX3CR1Ly108 subsets comprised between
10%–40% of the virus-specific CD8+ T cell compartment in the spleen, depending on the time of analysis (Figures 2B, 2C, and S4A). Given the differences in expression of chemokine receptors among these three subsets, we hypothesized that the frequency of each subset might vary by anatomical location. Notably, CX3CR1+, Ly108+, and CX3CR1Ly108 subsets were found to preferentially reside in distinct anatomical sites, with Ly108+ cells being widely restricted to secondary lymphoid tissues (Figures 2D and 2E). By contrast, the CX3CR1+ subset was the dominant population in the circulation, lungs, and inguinal lymph nodes, whereas the CX3CR1Ly108 subset was enriched in the bone marrow and the liver (Figures 2D and 2E). To determine whether gene expression of surface molecules and transcription factors that we observed in our scRNA-seq analyses correlated with protein expression, we co-stained LCMV-specific CD8+ T cells for CX3CR1, Ly108, and a representative set of markers. Consistent with our scRNA-seq data, we identified that the CX3CR1+ subset displayed increased expression of KLRG1, KLRa9, and T-bet, whereas Ly108+ cells exhibited high expression of CXCR5, CD127, and TCF-1 (Figures 2F–2I). Importantly, the CX3CR1Ly108 subset shared a similar expression profile to that of cluster 4 (Pdcd1+) cells, displayed elevated expression of CXCR6 and Eomes, and had increased expression of multiple co-inhibitory receptors, including PD-1, Tim3, and 2B4 (Figures 2F–2I). Ex vivo functional analyses of these 3 major subsets indicated that Ly108+ CD8+ T cells exhibited an enhanced capacity to co-produce interferon (IFN)-g and tumor necrosis factor alpha (TNF-a) upon GP33–41 peptide stimulation, whereas the CX3CR1Ly108 subset displayed the lowest potential to produce pro-inflammatory cytokines, consistent with their exhausted phenotype (Figures 2J and S4C). Notably, we further identified that CX3CR1+ CD8+ T cells displayed increased expression of granzyme B (Figure 2K), which correlated with their augmented cytotoxicity against peptide-pulsed target cells (Figure 2L). These data suggest that the CX3CR1+ subset has enhanced cytolytic function and may be a potent killer of virus-infected cells. Collectively, and in line with our scRNA-seq analyses, these findings demonstrate that differential expression of CX3CR1 and Ly108 can be used to distinguish 3 major subsets of CD8+ T cells during chronic viral infection, each of which displays distinct properties in their phenotype, transcriptional profile, anatomical distribution, and effector function.
Figure 2. Expression of Ly108 and CX3CR1 Defines Three Subsets of CD8+ T Cells during Chronic Viral Infection (A) t-SNE plot highlighting the three main subsets of CD8+ T cells present after establishment of chronic viral infection. (B) Representative flow plots depicting Ly108+, CX3CR1Ly108, and CX3CR1+ GP33+ splenic CD8 subsets on day 14 p.i. (C) Kinetics (mean ± SEM) of subsets in the spleen. Viremia (mean ± SEM) kinetics overlayed in shaded yellow is shown. (D) Representative flow plots depicting GP33+ CD8+ T cell subsets in indicated tissues on day 14 p.i. (E) Frequency of CD8+ T cell subsets in different anatomical locations. (F and G) Representative flow plots (F) and summary data (G) showing relative expression of surface molecules in GP33+ subsets on days 21–30 p.i. (H and I) Representative flow plots (H) and summary data (I) showing relative expression of TFs in GP33+ subsets. (J) Representative flow plots and summary data showing proportion of Ly108+, CX3CR1Ly108, and CX3CR1+ subsets degranulating (CD107a+) and producing IFN-g or co-producing TNF-a and IFN-g+ upon ex vivo GP33–41 stimulation. (K) Flow plot and summary data showing granzyme B expression in GP33+ subsets. (L) Summary data depicting relative cytotoxicity of Ly108+, CX3CR1 Ly108, and CX3CR1+ subsets against peptide-pulsed target EL4 cells. GP33-specific subsets were sort purified on day 21 p.i. and cultured with target cells at a 5:1 ratio. Data (mean ± SD in E, G, and I–K or mean ± SEM in L) are from 3–8 mice and are representative of at least 3 independent experiments.*p < 0.05, **p < 0.01, ***p < 0.001. See also Figure S4.
4 Immunity 53, 1–15, December 15, 2020
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
A
B Day 0: Infect CX3CR1DTR dLck-creor dLck-cre+ mice with LCMV Cl13
51.7
2.06
2.40
CR1DTR
CX3 dLck-cre -
DT injections on days 14, 16, 18, & 20
10
5
10
5
21.3
25.0
12.3
2.98
1.57
Harvest tissues for analyses on Day 23 post infection
10
4
10
CX3CR1
CX3CR1DTR dLck-cre +
Db-GP33
10
3
10
3
0
0
-10
4
3
-10
-10
3
0
10
3
10
4
10
3
63.9
5
-10
20.8
3
0
10
60 40 20 0 0
1
2
3
4
LCMV GP copy # (x105)
5
60 40 20 0 0.0
0.5
1.0
1.5
LCMV GP copy # (x106)
Cx3cr1+ CD8+ T Cells Are Required to Control Chronic Viral Infection As the CX3CR1+ CD8+ T cell subset displayed enhanced cytotoxicity ex vivo (Figure 2L), we next sought to determine whether CX3CR1+ CD8+ T cells play a critical role in controlling viral replication during persistent infection. To do this, we generated Cx3cr1DTR dLck-cre mice, which allows for the selective depletion of CX3CR1+ T cells in vivo upon administration of diphtheria toxin (DT) (Figure 3A; Zhang et al., 2005). As expected, upon sustained treatment with DT, we observed 55%–70% decreases in the proportion of circulating and splenic CX3CR1+GP33+ CD8+ T cells in LCMV Cl13-infected Cx3cr1DTR dLck-cre+ mice as compared to their Cx3cr1DTR dLck-cre- counterparts (Figures 3B and 3C). Notably, abrogation of the CX3CR1+ CD8+ T cell subset was further accompanied by approximately 4- to 5-fold increases in viral copy numbers detected in the blood and spleen, respectively (Figure 3D). Moreover, we identified that viral load significantly and inversely correlated with the magnitude of virus-specific CX3CR1+ CD8+ T cells (Figure 3E). Of note, we observed minimal expression of CX3CR1 on CD4+ T cells residing in either the blood or spleen, and treatment with DT did not result in any significant changes in the proportion of CX3CR1+ CD4+ T cells (Figure S5B). Thus, these data indicate that the impaired viral control observed in Cx3cr1DTR dLck-cre+ mice
re +
-
-c
re
ck
ck dL
P = 0.028 r square = 0.34
80
4
10
5
104
-c
re + -c
re -
ck
-c ck
-c ck
ck
dL
dL
P = 0.032 r square = 0.35
80
% CX3CR1+ of GP33+ (blood)
% CX3CR1+ of GP33+ (spleen)
E
10
(A) Experimental design. (B and C) Representative flow plots (B) and summary data (C) showing proportion of circulating or splenic CX3CR1+, Ly108+, and CX3CR1Ly108 CD8+ T cell subsets in Cx3cr1DTR dLck-cre or Cx3cr1DTR dLck-cre+ mice on day 23 p.i. (D) Summary data showing GP copy number in the sera or spleen of experimental mice on day 23 p.i. (E) Summary data showing inverse correlations between the proportion of CX3CR1+CD8+ T cells and viral burden in the spleen or blood. Data (mean ± SEM) in (C) and (D) are pooled, from 3–5 mice/group/experiment, and representative of at least 2 independent experiments.*p < 0.05, **p < 0.01, ***p < 0.001. See also Figure S5.
105
103
dL
re -c
re
dL
104
103
0
ck
ck
105
106
dL
20
LCMV copy # / mg spleen
40
-c
-c
60
+
0
**
106
LCMV copy # / mL sera
20
*
80
dL
% CX3CR1+ of GP33+ (spleen)
40
dL
D
**
60
re -
% CX3CR1+ of GP33+ (blood)
*** 80
re +
C
3
Ly108
CD44
Figure 3. CX3CR1+ CD8+ T Cells Are Critical for Control over Chronic Viral Infection
upon DT treatment was likely due to a loss in protective CX3CR1+ CD8+ T cells. Collectively, these data demonstrate a requirement for the CX3CR1+ CD8+ T cell subset in limiting viral replication during persistent viral infection.
Slamf6+ CD8+ T Cells Give Rise to Both Pdcd1+ Exhausted and Cx3cr1+ Effector CD8+ T Cell Subsets Recently, it has been proposed that CD8+ T cells responding to chronic viral infection exist as two major subsets: a CXCR5hiTCF-1hi progenitor population and a more terminally exhausted CXCR5loTCF-1lo population (He et al., 2016; Leong et al., 2016; Utzschneider et al., 2016; Im et al., 2016). However, after uncovering additional heterogeneity using scRNA-seq, we sought to determine the lineage relationship between the three major CD8+ T cell subsets that formed during persistent infection. To do this, we first used Monocle 2, which employs a machine-learning algorithm known as reverse graph embedding (RGE), to construct cell differentiation trajectories from our scRNA-seq data (Trapnell et al., 2014; Qiu et al., 2017). For this analysis, cells from the three major subsets present on day 30 p.i. (Slamf6+, Pdcd1+, and Cx3cr1+ cells), as well as clusters 3 (Slamf6+Pdcd1+) and 5 (Pdcd1+Cx3cr1+), which may represent intermediate states, were included (Figure 4A), and the top 100 marker genes for the Slamf6+, Pdcd1+, and Cx3cr1+ clusters were used to construct the trajectory. We found that the algorithm predicted a differentiation trajectory with one major branch point, in which Slamf6+ cells could form both Pdcd1+ and Cx3cr1+ cells as terminal endpoints (Figure 4A). In this trajectory, cluster 3 (Slamf6+Pdcd1+) cells were found mostly between Slamf6+ cells and the branch point and also between the branch point and Pdcd1+ cells. Conversely, cluster 5 (Pdcd1+Cx3cr1+) cells were found mostly between the branch point and the Pdcd1+ and Cx3cr1+ subsets (Figure 4A). As expected, when visualizing expression of marker genes for the three major subsets over the course of Immunity 53, 1–15, December 15, 2020 5
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
B
C
Cluster 2 markers
A
Cluster 4 markers
Day 0 Infect WT B6 mice with LCMV Cl13
Cluster 6 markers
Day 45: Sort and transfer 60K CX3CR1+, Ly108+, and CX3CR1-Ly108- CD8+ T cell subsets into naive Ly5.1 mice and challenge with LCMV Cl13
Brdu labeling and collect blood and tissues for analysis
Pseudotime
D
E
Blood 93.8
1.43
Spleen
2.2
Liver
0.4
86.3
0.4
13.2
0.13
1.2
48.6
2.5
45.9
2.9
0.8
81.9
9.3
9.5
0.3
89.5
0.7
56.1
0
0
AT CX3CR1+
3.57
3.6
0.3
41.1
3.5
47.2
8.2
59.0
0
18.1
0
AT Ly108+
10
10
2
10
4
10
60
5
CX3CR1
0
-10
0
3
-10
3
0
10
4
10
5
10
3
0
2
0
102
Ly108
103
104
3
42.1
1.8
3
105
-10
3
0
Ly5.1
4
10
5
10
10
Ly108
20
**** -
0 08
0
40
Ly 1
20
08 +
40
60
R 1-
X 3C C
60
** *
** *
80
X 3C
0
** **
80
C
*** ***
100
Ly 1
*** ***
50
** **
100
Subset distribution of Donor CD8+ T cells in Liver Day 21 (%)
H
C X 3C R 1+
ve r Li
Sp l
ee
n
0
3
Subset distribution of Donor CD8+ T clells in Spleen Day 21 (%)
2
10
*** ***
100
08 +
* **
10
Ly5.1
Ly 1
** ***
3
-10
0
3
10
Ly5.2
0
5
10
1+
12
od
4
10
G ** **
Bl o
3
10
R
% Ly5.2+ Donor of CD44hi CD8+ T cells
0
Ly108
16
4
CX3CR1
Ly5.2 3
-10
AT CX3CR1+ AT Ly108+ AT CX3CR1-Ly108-
8
0
41.0
5
10
10
0
8-
4
10
C X 3C
3
10
Ly5.1
Subset distribution of Donor CD8+ T cells in blood Day 9 (%)
0
4
3
10
Ly 10
CX3CR1
Ly5.2
3
-10
3
-10
5
10
10
3
102
0
3
0.16
105
104
4
3
10
-10
0
10
10 3
0
40
4
10
10
F
5
10
4
10
0.08
5
Ly 10 C X 8+ 3C R 1Ly 10 8-
4
10
1+
10
R 1-
AT CX3CR1Ly108-
5
0.30
C X 3C R
5
10
Figure 4. Lineage Relationship between CD8+ T Cell Subsets (A) Single cell trajectory of CD8+ T cell subsets present on day 30 p.i. (clusters 2–6). (B) Heatmap showing expression of clusters 2, 4, and 6 markers used for trajectory analysis over pseudotime. (C–H) Experimental design (C) 60,000 CD45.2+CD44hi CD8 T cells from each subset (Ly108+, CX3CR1 Ly108, and CX3CR1+) were transferred to CD45.1recipient mice. These mice were then infected with LCMV Cl13 1 day later. (D and E) Representative flow plots showing expansion (left) and phenotype (right) of cells from the blood on day 9 p.i. (D) or spleen and liver (E) on day 21 p.i. (F–H) Summary of transferred cell frequency (F) and phenotype (G and H) in either the blood (G) or spleen and liver (H). Data (mean ± SD in F–H) are from 3 to 4 mice/group and are representative of 3 independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001. See also Figure S6.
6 Immunity 53, 1–15, December 15, 2020
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
pseudotime, a measure of progress through the differentiation trajectory, we found that genes expressed by both Pdcd1+ and Cx3cr1+ CD8+ T cells appeared later than those expressed by Slamf6+ cells, suggesting that Pdcd1+ and Cx3cr1+ effector subsets represent more terminally differentiated populations (Figure 4B). Next, we scored each cell based on the overall expression of the top 100 marker genes for the Slamf6+, Pdcd1+, and Cx3cr1+ CD8+ T cells to determine whether clusters 3 and 5 may, in fact, be intermediate states in the differentiation process. We found that Slamf6+ markers were also highly expressed by most cluster 3 (Slamf6+Pdcd1+) cells, although a small subset of cluster 3 cells had already turned down expression of these genes (Figure S6A). Additionally, expression of genes enriched in Pdcd1+ cells was turned on in a small subset of cluster 3 cells, and cluster 5 (Pdcd1+Cx3cr1+) cells expressed an intermediate level of these genes (Figure S6A, middle). Markers of Cx3cr1+ cells were also intermediately expressed in cluster 5 cells (Figure S6A, bottom). Collectively, our scRNAseq trajectory analyses support a model in which Slamf6+ CD8+ T cells can differentiate into either Pdcd1+ or Cx3cr1+ cells, with clusters 3 (Slamf6+Pdcd1+) and 5 (Pdcd1+Cx3cr1+) cells potentially existing as transition states between these three subsets. To determine the in vivo differentiation trajectory, proliferative potential, and phenotypic stability of the CX3CR1+, Ly108+, and CX3CR1Ly108 (PD-1hi) subsets, we performed adoptive transfer experiments using congenically marked CD8+ T cells. To do this, we sort-purified effector CD8+ T cell subsets from infected CD45.2+ donor mice and transferred equal numbers of cells to naive CD45.1+ mice, which were then subsequently challenged with LCMV Cl13 1 day later (Figures 4C and S6B). Notably, compared to CX3CR1Ly108 cells or CX3CR1+ cells, we found that Ly108+ cells displayed vigorous expansion in the blood, spleen, and liver (Figures 4D–4F), which was associated with their increased expression of Ki67 and incorporation of bromodeoxyuridine (BrdU) relative to the CX3CR1+ subset (Figure S6C). Moreover, and consistent with our Monocle 2 predictions, Ly108+ CD8 T cells were found to give rise to both CX3CR1+ and CX3CR1Ly108 subsets (Figures 4D, 4E, 4G, and 4H), further supporting the identity of this subset as the recently described progenitor population with stem-cell-like characteristics (He et al., 2016; Im et al., 2016; Leong et al., 2016; Utzschneider et al., 2016). We also identified that CX3CR1+ cells accumulated to a greater extent than CX3CR1Ly108 donor cells in all tissues assessed (Figures 4D–4F), although this did not reach the level of statistical significance. Importantly, the majority of donor-derived cells from the CX3CR1+ subset retained high CX3CR1 and T-bet expression and did not differentiate into Ly108+ or CX3CR1Ly108 CD8+ T cells (Figures 4D, 4E, 4G, 4H, and S6D), further indicating their terminal differentiation status. By contrast, although CX3CR1Ly108 (PD-1hi) donor cells accumulated to the lowest extent, approximately 40%–50% of their recovered progeny acquired high CX3CR1 and T-bet expression (Figures 4D, 4E, 4G, 4H, and S6D). This observation could potentially be explained by additional heterogeneity residing among the CX3CR1Ly108 population, such as their encompassing either Slamf6+Pdcd1+ or Pdcd1+Cx3cr1+ intermediate populations (clusters 3 and 5; shown in Figure 1), which are in transitional differentiation states and which may
have distinct developmental potential from the PD-1hi terminally exhausted subset. Overall, our data are likely suggestive of a bifurcation model wherein Ly108+ cells can directly form either CX3CR1Ly108 or CX3CR1+ cells, with a small fraction of cells within the CX3CR1Ly108 subset maintaining their potential to develop into CX3CR1+ cells. CD4+ T Cell Help via IL-21 Is Critical for the Generation of the CX3CR1+ CD8+ T Cell Subset Given this unexpected heterogeneity in the antiviral CD8+ T cell pool, coupled to the critical role for CD4+ T cell help in sustaining CD8+ T cell responses during persistent viral infection, we next sought to determine whether CD4+ T cell help regulates this multifaceted process of CD8+ T cell differentiation. Notably, CD8+ T cell intrinsic IL-21R signaling has previously been demonstrated to be essential for sustaining CD8+ T cell responses during chronic viral infection, and CD4+ T cells are implicated as being the major source of IL-21 (Elsaesser et al., 2009; Fro¨hlich et al., 2009; Xin et al., 2015). In line with this, our data identified that CD4+ T cells comprised over 90% of IL-21producing lymphocytes in the spleen during LCMV Cl13 infection (Figure S7A). Nevertheless, how CD4+ T-cell-derived IL-21 and CD8+ T-cell-intrinsic IL-21R signaling regulate CD8+ T cell differentiation during chronic viral infection remains unclear. To gain insight into how the IL-21R signaling pathway may impact CD8+ T cell differentiation during chronic viral infection, we first compared the relative expression of Il21r on the three main day 30 LCMV-specific effector subsets. At least moderate amounts of Il21r expression could be detected in each CD8+ T cell subset (Figure 5A), although our scRNA-seq analyses identified that CX3CR1Ly108 (PD-1hi) cells displayed the highest expression of Il21r, possibly suggesting a role for CD4+ T cell help in regulating the differentiation of this phenotypically and functionally exhausted subset. To test this hypothesis, we employed three different experimental approaches. First, we used a transient CD4+ T-cell-depletion model that is known to result in sustained viremia and severe CD8+ T cell dysfunction during chronic LCMV infection (Battegay et al., 1994; Matloubian et al., 1994; Zajac et al., 1998). As expected, mice depleted of CD4+ T cells 1 day before LCMV Cl13 infection exhibited significantly impaired viral control (Figure S7B). Notably, depletion of CD4+ T cells resulted in 50%–75% decreases in the proportion of CX3CR1+ LCMV-specific CD8+ T cells circulating in the blood and inhabiting the spleen (Figures 5B and 5C). Although this decrease in the CX3CR1+ subset was accompanied predominantly by an increase in the CX3CR1Ly108 subset in the blood, the frequency of both Ly108 and CX3CR1Ly108 subsets was significantly increased in the spleen in the absence of CD4+ T cell help (Figures 5B and 5C). These differences in the subset distribution of LCMV-specific CD8+ T cells were further associated with markedly reduced expression amounts of T-bet to Eomes (Figure S7E), indicating that changes in the overall transcriptional program may be responsible for these alterations in subset skewing. Second, we used a co-adoptive transfer model wherein wild-type (WT) and Il21r/ P14 cells, which express a transgenic T cell receptor (TCR) specific for GP33–41 (Pircher et al., 1990), were transferred into congenically marked recipient mice at a 1:1 ratio before initiating LCMV Cl13 infection the next day (Figures 5D and S7F–S7I). Notably, use of this experimental design Immunity 53, 1–15, December 15, 2020 7
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
A
B Blood
Spleen
83.9
4.4
10.7
71.4
2.6
12.6
13.3
7.87
Control
Il21r 5 10
10
5
3.2
8.5
15.8
0
10
5
5
10
8.9
1.8
4.27
10
0
10
3
10
0
3 -10
10
3
10
4
10
3
6.8
77.4
5 -10
3
0
CD44
C
3
3
10
4
10
-10
3
10
0
3
-10
5
-10
Ly108
3
0
10
3
10
4
10
CD44
3
61.0
5
-10
3
28.3 0
10
3
4
10
10
Ly108
5
D
-10
0
10
3
Ly5.1
10
4
10
10 -10
3
0
10
3
10
4
Ly5.2
10
5
3
20 0
0
-10
+
3
5
4
08
-10
3
10
40
3
20.0 -10
3
55.0 0
10
3
10
4
10
5
R
3 -10
IL-21R-/10.1
0
3.30
X 3C
0
3
6.11
21.7
R
10
Thy1.1
Ly5.2 (P14)
3 10
5
1Ly 10 8-
27.5
CX3CR1
R X 3C C
WT 87.9
4
10
0
08
5
C
20
10
1+
10
4 10
***
60
X 3C
40
***
80
Ly 1
Gated on Ly5.2 P14
Donor 17.7
5 10
+
100
C
60
-
+
C
X 3C
R
1Ly 1
08 Ly 1
C
X 3C
R
1+
0
2.16
Analyze P14 response Day 21 post-infection Gated on CD8+ CD44hi
-
20
***
08 + X 3C R 1Ly 10 8
*
40
**
64.2
C
60
***
1+
80
80
Ly 1
***
Subset distributionof GP33+ CD8+ T cells in spleen (%)
***
100
WT -/Il21r
Day 0 Infect with LCMV Cl13
P14 subset distribution (spleen) (%)
Day -1 AT Thy1.1 WT & Ly5.2 Il21r-/- P14 cells into Ly5.1 recipient mice at a 1:1 ratio
Control α-CD4 Subset distributionof GP33+ CD8+ T cells in blood (%)
10
4
10
0
-10 0
4.75
4
CX3CR1
3 10
4
Db-GP33
10
CX3CR1
Db-GP33
α-CD4
4 10
Ly108
Figure 5. CD4+ T Cell Help via IL-21 Is Critical for the Generation of CX3CR1+ CD8+ T Cells (A) t-SNE plot or violin plots highlighting the three major subsets of CD8 T cells during chronic LCMV infection and their relative expression of Il21r. (B and C) Representative flow plots (B) and summary data (C) showing the CX3CR1+, Ly108+, or CX3CR1Ly108 subset distribution of GP33+CD8 T cells in the blood or spleen of control or CD4-depleted mice on day 21 p.i. (D) Experimental design (top), representative flow plots (bottom), and summary data (right) depicting the subset distribution of WT and Il21r/ P14 cells on day 21 p.i. Data (mean ± SEM) in (C) and (D) are pooled, from 3–5 mice/group/experiment, and representative of at least 3 independent experiments.*p < 0.05, **p < 0.01, ***p < 0.001. See also Figure S7.
helps control for WT and Il21r/ P14 cells to experience a similar degree of exposure to both viral load and inflammation in the same host. Importantly, deletion of IL-21R signaling in P14 transgenic CD8+ T cells strongly abrogated the development of the CX3CR1+ CD8+ T cell subset, as witnessed by 75% decreases in the frequency of splenic CX3CR1+ P14 cells (Figure 5D). In accord with our CD4+ T cell depletion experiments, we observed substantial reductions in the relative expression of T-bet to Eomes in Il21r/ P14 cells (Figure S7H). Of note, however, despite this skewing in CD8+ T cell subset distribution, we observed a similar capacity between WT and IL-21R-deficient P14 cells to degranulate and produce IFN-g upon ex vivo peptide stimulation (Figure S7I). This may possibly be due to IL-21R-deficient P14 cells retaining a high proportion of Ly108+ progenitor CD8+ T cells, which we demonstrate exhibit a high capacity to degranulate and produce effector cytokines upon ex vivo peptide stimulation (Figure 2). Lastly, to assess whether CD8+ T-cell-intrinsic IL-21 signaling was important for the formation of virus-specific CX3CR1+ effector cells originating from a poly8 Immunity 53, 1–15, December 15, 2020
clonal repertoire, we generated WT and Il21r/ CD8+ T cell mixed bone marrow chimera mice (Figure S7J) and challenged them with LCMV Cl13. Notably, and consistent with our P14 adoptive transfer experiments, abrogation of IL-21 signaling in the CD8+ T cell compartment resulted in a severe impairment in the development of GP33-41+ CX3CR1+ CD8+ T cells during chronic LCMV infection (Figure S7K), which was further associated with decreased control over viral replication (Figure S7L). Taken together, these data indicate a critical role for CD4+ T-cell-derived IL-21 in promoting the formation of CX3CR1+ cytotoxic CD8+ T cells, likely by facilitating the differentiation of Ly108+ progenitor cells along a CX3CR1+ developmental pathway, while simultaneously limiting CD8+ T cell differentiation toward the PD-1hi exhausted subset. PD-L1 Blockade Is Insufficient to Rescue CX3CR1+ Differentiation among Un-helped CD8+ T Cells Given the extreme defect in the formation of the CX3CR1+ subset in the absence of CD4+ T cell help and the ensuing increase in the
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
C
64.8
4.5
GP33 (%)
16.5
*
6 4
4.7
2
+ α-PD-L1
0
1+
4 αα - CD PD 4 -L + 1
R
1
20
C X 3C %
α-
C
-L
tro on
PD
0.6
α-
15.4
C
15.3 9.8
40
D
l
0
60
1.4
5.5
21.8
0
15.2
α-CD4 6.09
7.60
α-CD4 + α-PD-L1 1.37
9.79
3.94
4
3
0
85.8 -10
3
2.14 0
10
3
10
4
10
76.7
2.01
89.5
1.56
84.2
5
2 0
C
IFN-γ
2.07
4
l
3
**
6
tro
-10
8
** ** *** ***
107 106 105 104
10
1
αC D 4 αα- CD PD 4 -L + 1
10
3.47
on
CD107a
10
8.55
* ***
10
G
l
+ α-PD-L1
Control 5
*
1
F
10
0
5
-L
10
tro
4
on
10
PD
3
Ly108
C
10
α-
0
g3
3
La
-10
Serum viral load (PFU/mL)
72.3
3
5
Ti m
10
-1
4
PD
10
500
-L 1 αC D 4 αα- CD PD 4 + -L 1
3
*** *** ***
1000
PD
10
CD44
4000
α-
0
C
3
CD107a+ IFNγ+ CD8+ Tcell upon GP33 stim (%)
-10
-10
3
Control α-PD-L1 α-CD4 α-CD4 + α-PD-L1
8000
4 αα- CD 4 PD -L + 1
3
1
l
-10
3
0
1
10
0
*
2
**
D
3
4
tro
10
10
on
Db-GP33
α-CD4 + α-PD-L1
4
CX3CR1
10
12000
gMFI co-inhibitory receptor
4.4
*** ** ***
E
αC
5
-L
10
*
3
D
5
30.7
*** ***
αP
10
59
ratio Tbet to Eomes gMFI
D
%
2.0
α-CD4
Control α-PD-L1 α-CD4 α-CD4 + α-PD-L1
1Ly 10 8-
** ***
8 23.2
*** *** ** **
** **
R
Control
*** *** *** ***
80
8+
***
10
Ly 10
**
4.2
C X 3C
2.9
%
B 57.4
CD8+ T cell subset distribution (%)
A
Figure 6. Blockade of PD-1-PD-L1 Axis Is Insufficient to Rescue the CX3CR1-Differentiation Defect of Un-helped CD8+ T Cells (A–F) Separate groups of control or CD4-depleted mice were infected with LCMV Cl13 and treated with isotype or anti-PD-L1 antibodies on days 16, 19, and 22 p.i. GP33+CD8 T cell responses were assessed on day 23 p.i. (A–C) Representative flow plots (A) and summary data (B and C) showing the proportion of splenic CD8 T cells (B) and their subset distribution (C). (D) Relative expression geometric mean fluorescence intensity (gMFI) of T-bet to Eomes in GP33+CD8+ T cells from indicated experimental groups. (E) Summary graph showing the relative expression of inhibitory molecules on GP33+ cells from experimental mice. (F) Representative flow plots and summary data depicting the proportion of CD107a+IFN-g+CD8 T cells upon ex vivo GP33–41 stimulation. (G) Summary data showing viremia in experimental mice on day 23 p.i. Data (mean ± SEM) in (B)–(G) are pooled, from 3 mice/group/experiment, and representative of at least 2 independent experiments.*p < 0.05, **p < 0.01, ***p < 0.001.
proportion of CX3CR1Ly108 exhausted cells, we next questioned whether PD-L1 blockade, an immunotherapy known to partially reverse T cell exhaustion during persistent infection and cancer (Barber et al., 2006; Wherry and Kurachi, 2015), could rescue the development of the CX3CR1+ subset in LCMV Cl13-infected mice. Notably, although PD-L1 blockade in CD4+ T-cell-depleted mice augmented both the magnitude of the GP33–41+ CD8+ T cell response and the capacity of LCMV-specific cells to degranulate and secrete IFN-g, it was not sufficient to rescue the formation of the CX3CR1+ subset (Figures 6A–6C and 6F). Similarly, a-PD-L1 treatment in CD4+ T-cell-intact mice also had no appreciable effect on the subset distribution
of GP33–41+ CD8+ T cells (Figures 6A and 6C), despite resulting in an 80%–90% reduction in viremia compared to control mice (Figure 6G). Additionally, despite displaying enhanced accumulation and augmented cytokine production, the majority of LCMV-specific CD8+ T cells from combinatorial a-CD4 and a-PD-L1-treated mice were actually found to reside within the CX3CR1Ly108 subset and coordinately expressed increased amounts of PD-1 and Tim3 and diminished T-bet to Eomes amounts as compared to all other experimental groups (Figures 6A, 6D, and 6E). Moreover, a-PD-L1 treatment (beginning week 3 of LCMV Cl13 infection) in CD4+ T-cell-depleted mice had only a minor (and non-statistically significant) impact on viral control Immunity 53, 1–15, December 15, 2020 9
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
B
Gated on CD44hi PD-1hi CD8+ TILS 20.7
0.4
80 10
4
3
40 2
20
0
C
7.9 10
2
10
3
10
Ly108
4
10
17
5
0 0
10
100
80 5
10
0.63
63
1
4
10
CX3CR1
10
3
10
4
PD-1
10
0
5
2.2
ACT IL-21- CD4+ T cells
ACT IL-21+ CD4+ T cells
2
1000
3
10
0
36
0.09 0 10
3
4
10
10
D **
2.0
100
1.0
40
0.5
20
0.0
0
CD AC 4 + T IL T -21 ce lls A CD CT 4 + IL T -21 + ce lls
E
Assess T cell response 8 Days post ACT
1.5
60
5
Ly108
**
80
Measure tumor volume
C X C 3C X R 3C 1+ R 1Ly 10 8-
71
+ ACT of IL-21-tRFP or IL-21-tRFP + CD4 T cells activated with tumor-pulsed DCs under Th17 conditions
CX3CR1+ of CD8+ TILs (x103)
0
7-10 Days
2000
r = -0.7345 P = 0.0003
80 60 40 20
CD AC 4 + T IL T -21 ce lls CD ACT 4 + IL T -21 + ce lls
10
CX3CR1+ of CD8+ TILs (%)
CX3CR1
60 10
Inject B16-F10 melanoma s.c.
100
Tumor volume (mm3)
5
gMFI of PD-1
10
*
3000
CX3CR1+ of CD8+ TILs (%)
A
0 100
101
102
Tumor size
150
IL-21- CD4+ T cells IL-21+ CD4+ T cells
*
100 50 0
103
(mm3)
0
2
4
6
8
Days after ACT
11
ACT IL-21- CD4+ T cells
0
0 100
101
102
103
Tumor size (mm3)
G
H 25
30%
cd8-/-
+ or +
lethally irradiated
WT
WT
WT
30% il21r-/-
8 weeks 5
10
12
12
0
4
7-10 Days + ACT of IL-21-tRFP + CD4 T cells activated with tumor-pulsed DCs under Th17 conditions
20 10 0
10
2
60
30
3
10 0
40
**
W
Il21r -/-
20
40
10
CX3CR1
Inject B16-F10 melanoma s.c.
60
0
80
100
CX3CR1+ of CD8+ TILs (%)
T
70%
3
0
*
1.5
1.0
0.5
0.0 -/-
5
10
T
4
10
W
3
10
CD44
CD AC 4 + T IL T -21 ce lls CD ACT 4 + IL T -21 + ce lls
0
20
21 r
0
40
IIl
20
60
CX3CR1+ of CD8+ TILs (x103)
40
80
-/-
20
60
Il2 1r
40
80
CX3CR1+ of CD8+ TILs (%)
60
r = 0.8364 P < 0.0001
100
CX3CR1+ of CD8+ TILs (%)
3
10
80
r = -0.729 P = 0.0003
100
IL-21t-RFP+ of CD4+ T cells (%)
4
10
IL-21-tRFP
ACT IL-21+ CD4+ T cells
67
IL-21t-RFP+ of CD4+ T cells (%)
5
10
***
IL-21t-RFP+ of CD4+ T cells (%)
F 100
40
5
83 0
10
3
10
Ly108
4
10
5
r = -0.60 P = 0.03
30 20
Assess T cell response 8 Days post ACT
10 0 100
101
102
103
Tumor size (mm3)
Figure 7. Provision of IL-21+CD4 Help Enhances CX3CR1+ CD8+ T Cell Development and Control over Tumorigenesis
(A) Representative flow plots showing the %CX3CR1+, Ly108+, or CX3CR1Ly108 CD44hiCD8+ TILs responding to B16-F10 tumor and their relative expression of PD-1. (B) Experimental design. (C) Representative flow plots and summary data displaying the proportion and total number of CX3CR1+ TILs on day 8 post-ACT. Summary data show negative correlation (far right) between %CX3CR1+ TILs and tumor burden.
(legend continued on next page)
10 Immunity 53, 1–15, December 15, 2020
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
(Figure 6G), bringing to question the efficacy and sustainability of PD-L1 checkpoint blockade in the absence of CD4+ T cell help (and consequently the CX3CR1+ subset). Taken together, these data indicate that the PD-1 inhibitory pathway is unlikely to play a major role in regulating the differentiation of CX3CR1+ CD8+ T cells during chronic viral infection. Moreover, these data further demonstrate that blockade of the PD-1-PD-L1 axis is insufficient to rescue the CX3CR1-differentiation defect of ‘‘un-helped’’ CD8+ T cells. Provision of CD4+ T Cell Help in the Form of IL-21 Enhances CX3CR1+ CD8+ T Cell Formation and Tumor Control Lastly, given the striking resemblance between CD8+ T cells responding to chronic viral infection and tumor-infiltrating lymphocytes (TILs) (Ahmadzadeh et al., 2009; Barber et al., 2006; Blackburn et al., 2009; Sakuishi et al., 2010), we next sought to investigate whether CD8+ T cells invading the tumor microenvironment also displayed a similar pattern of differentiation as virus-specific cells during LCMV Cl13 infection. To do this, we employed the well-characterized B16-F10 melanoma model (Overwijk and Restifo, 2001), which is known to result in an extreme form of T cell dysfunction and for which current immunotherapy strategies are rather ineffective (Overwijk et al., 2003; Wang et al., 1998; Ahmadzadeh et al., 2009; Moroz et al., 2004). Importantly, even after preconditioning with sublethal irradiation (Klebanoff et al., 2005; Gattinoni et al., 2005; Hanada et al., 2019), the majority of CD8+ TILs lacked expression of both CX3CR1 and Ly108 (Figure 7A), consistent with a high level of T cell exhaustion occurring in the tumor microenvironment. Moreover, and similar to that observed in our persistent infection model, CX3CR1Ly108 cells displayed uniformly high expression of PD-1 (Figure 7A). Of note, we detected minimal Ly108+ cells in the tumor, possibly due to their preferential residence in secondary lymphoid tissues (Figure 2). Previous reports have identified that adoptive cell therapy (ACT) with CD4+ T cells can improve CD8+ T cell responses and enhance control over tumor progression (Lu et al., 2014; Muranski et al., 2008; Purwar et al., 2012; Ve´gran et al., 2014; Søndergaard et al., 2010). Many of these studies have further identified that the therapeutic benefit of this CD4+ T-cell-mediated therapy is associated with CD4+ T cell secretion of IL-21 (Moroz et al., 2004; Ve´gran et al., 2014; Søndergaard et al., 2010), although it remains unclear how CD4+ T cell help regulates the differentiation trajectory of CD8+ T cells. Thus, we next asked whether provision of IL-21 via CD4+ T cell help could enhance the development of the CX3CR1+ subset and its infiltration into the tumor and whether this could confer protection against tumor growth. To generate tumor-specific IL-21+ CD4+ T cells, we used an
in vitro activation approach wherein CD4+ T cells from IL-21 turbo red fluorescent protein (IL-21-tRFP) reporter mice (Shulman et al., 2014) were primed with tumor-pulsed dendritic cells (DCs) under T helper-17 (Th17) cell conditions. Next, IL-21tRFP and IL-21-tRFP+ Th17 cells were sort purified and ACT was administered to mice that were previously injected with B16-F10 melanoma cells 7–10 days prior (Figure 7B). We then proceeded to measure tumor burden in these experimental mice over time, and after 8 days post-ACT, we assessed polyclonal T cell responses in the tumor. Remarkably, recipients of IL-21-tRFP+ CD4+ T cells displayed over a 2-fold increase in the proportion of tumor-infiltrating CX3CR1+ CD8+ T cells, which was highly associated with their enhanced control over tumor growth (Figures 7C and 7D). Moreover, the proportion of CX3CR1+ CD8+ T cells strongly correlated with the magnitude of the IL-21+ CD4+ T cell helper response, which in turn was also found to inversely correlate with tumor burden (Figures 7E and 7F). Thus, our data indicate that provision of IL-21+ CD4+ T cell help in the form of ACT is sufficient to augment the expansion of CX3CR1+ CD8+ TILs, which is mechanistically linked to improved tumor control. To determine whether the observed enhancement in CX3CR1+ CD8+ TIL formation and tumor control afforded by IL-21+ CD4+ T cell ACT is dependent on direct IL-21 signaling in CD8+ T cells, we generated WT and Il21r/ CD8+ T cell mixed bone marrow chimera mice and repeated our B16-F10 melanoma experiments (Figure 7G). Importantly, the expansion of CX3CR1+ CD8+ TILs upon treatment with IL-21+ CD4 ACT was largely abolished when IL-21 signaling in CD8+ T cells was abrogated (Figure 7H). Additionally, the magnitude of CX3CR1+ CD8+ TILs once again inversely correlated with tumor burden (Figure 7H), indicating that CD8+ T cell intrinsic IL-21 signaling is required for the therapeutic efficacy of IL-21+ CD4+ T cell ACT. Taken together, our findings demonstrate that CD4+ T cell help plays a critical role in the generation of protective cytotoxic CX3CR1+ CD8+ T cells and that this differentiation checkpoint may potentially be exploited for the purpose of ACT. DISCUSSION Collectively, our work has revealed the development of three major CD8+ T cell subsets during persistent viral infection, with each subset differing in their transcriptional program, phenotype, functional features, and tissue distribution. Thus, our findings add an additional layer of complexity to the previously proposed model of CD8+ T cell differentiation during chronic infection. Additionally, our study highlights a critical role for CD4+ T cell help via IL-21 in the generation of a transcriptionally and functionally distinct CX3CR1+ cytotoxic CD8+ T cell subset that is
(D) Tumor growth curves; of note, summary data are from (n = 6) IL-21 Th17 cell recipients and (n = 8) IL-21+ Th17 cell recipients. IL-21 Th17-cell-recipient mice that developed >30% of an IL-21+tRFP+ response (n = 3) were excluded from the analysis. (E) Representative flow plots and summary data displaying the %IL-21-tRFP+ CD4 T cells in the tumor at day 8 post-ACT. Summary data (right) show inverse correlation between IL-21-tRFP+ CD4 T cells and tumor size. (F) Summary data show positive correlation between IL-21+CD4+ T cell response and %CX3CR1+CD8+ TILs. (G) Experimental design. (H) Representative flow plots and summary data displaying the proportion and total number of CX3CR1+CD8+ TILs on day 8 post-ACT. Summary data (bottom) show inverse correlation between %CX3CR1+ TILs and tumor burden. Data (mean ± SEM) in (C)–(E) and (H) are pooled, from 3–5 mice/group/experiment, and representative of at least 2 independent experiments.*p < 0.05, **p < 0.01, ***p < 0.001.
Immunity 53, 1–15, December 15, 2020 11
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
essential for viral control. Thus, our study addresses a longstanding question within the field and identifies a previously unrecognized mechanism by which CD4+ T cell help sustains protective antiviral CD8+ T cell responses during chronic viral infection. Overall, our data indicate that the self-renewing progenitor population developed early and remained in a transcriptionally similar state during the later phase of chronic viral infection. As the infection progressed, these Slamf6+ progenitor cells could potentially give rise to two distinct terminally differentiated effector CD8+ T cell subsets: a Pdcd1+ population and a CX3CR1+ subset. These two late-phase effector subsets displayed unique properties, with Pdcd1+ cells appearing to be highly dysfunctional, as exemplified by their poor recall potential and diminished cytokine production and cytolytic activity. Conversely, Cx3cr1+ cells exhibited markedly superior cytolytic function and a heightened capacity to degranulate and secrete effector cytokines. This transcriptional switch, from the bifurcation of Slamf6+ progenitor cells into either the Pdcd1+ or Cx3cr1+ cell subsets, likely marks the start of the true chronic infection differentiation program. Moreover, we speculate that this division of labor is likely essential for maintaining viral control, while simultaneously limiting excessive immunopathology. T cell exhaustion is classically defined by sustained expression of co-inhibitory receptors, poor effector function, and a transcriptional state that is distinct from functional effector or memory T cells (Wherry and Kurachi, 2015; Wherry et al., 2003; Kahan et al., 2015). Additionally, exhausted T cells are implicated as being terminally differentiated (Wherry and Kurachi, 2015). In line with these studies, our scRNA-seq analyses and functional characterization of LCMV-specific CD8+ T cells identified a major population of CX3CR1Ly108 (PD-1hi) cells that appear highly dysfunctional and exhibit an exhausted phenotype. Moreover, our adoptive transfer experiments demonstrate that this CX3CR1Ly108 CD8+ T cell subset displays a reduced capacity to expand upon secondary recall. However, our recall experiments also demonstrated that some CX3CR1Ly108 cells do retain their capacity to differentiate into the CX3CR1+ subset. This observation could potentially be explained by the possibility that CX3CR1Ly108 cells may also contain Slamf6+Pdcd1+ or Pdcd1+Cx3cr1+ intermediate populations (clusters 3 and 5, respectively; Figure 1) that could have distinct recall properties. Alternatively, although unlikely, it may also be possible that some exhausted CD8+ T cells may retain the potential to redirect their differentiation toward the CX3CR1+ cytolytic subset. By contrast, the majority of CX3CR1+ donor-derived cells retained high CX3CR1 and T-bet expression upon recall, further supporting that the CX3CR1+ subset has progressed to a terminal state of differentiation. In an independent study, Hudson et al., 2019 [this issue of Immunity] have shown that CD101Tim3+ CD8+ T cells displayed a striking similarity to the CX3CR1+ subset identified in our study, as exemplified by their increased surface expression of CX3CR1, KLRG1, and the transcription factors T-bet, Zeb-2, and Klf-2. Consistent with their enhanced effector-like profile, CD101Tim3+ cells also displayed augmented IFN-g production and granzyme B expression relative to the exhausted CD101+ Tim3+ subset. Importantly, Hudson et al., 2019 similarly observed a critical role for CX3CR1+ (CD101Tim3+) CD8+ T cells in mediating antiviral immunity during chronic viral infection, thereby 12 Immunity 53, 1–15, December 15, 2020
underscoring the importance of maintaining this cytolytic effector subset. Of note, although some small differences in the expression pattern of certain molecules (such as Tim3 and Eomes) and in the responsiveness to PD-1 blockade were observed when comparing effector versus exhausted CD8+ T cell subsets in these two studies, respectively, this is likely a result of the stark differences in viral load that occurs following chronic infection under CD4+ T-cell-sufficient versus CD4+ T cell un-helped conditions. Additionally, although our study demonstrates that CX3CR1+ CD8+ T cells (isolated at the late phase of chronic infection when viral titers decline) appear terminally differentiated upon recall with LCMV Cl13 in naive recipient mice, Hudson et al., 2019 demonstrate that un-helped CD101Tim3+ cells adopt a transitory differentiation state and maintain their proliferative potential when transferred into infection-matched mice (where viral load remains high). In response to continuous antigen stimulation, CD101Tim3+ cells eventually progress into CD101+Tim3+ exhausted cells. Future work will be important for further dissecting the overall similarities and differences between CD101Tim3+ and CX3CR1+Ly108 effector subsets and determining how viral burden and CD4+ T cell help impacts their differentiation potential and function. Although previous studies have identified an essential role for IL-21 in sustaining virus-specific CD8+ T cell responses and viral control (Fro¨hlich et al., 2009; Tian et al., 2016; Yi et al., 2009), the mechanisms by which IL-21 confers this protection has thus far remained elusive. Importantly, our study identified that the generation of cytotoxic CX3CR1+ CD8+ T cells, which are essential for viral control, was critically dependent on CD4+ T cell help and CD8+ T cell intrinsic IL-21R signaling. Our study further indicated that CD4+ T cells comprise approximately 90% of all IL-21-producing lymphocytes responding to LCMV Cl13 infection, suggesting that T helper cells are likely the major source of IL-21 that drives the differentiation of CX3CR1+ CD8+ T cells. Importantly, our findings further identified that the CD4-IL-21CX3CR1 developmental pathway was also operational during tumorigenesis and may be harnessed to enhance immune-mediated control over tumor progression. Despite these findings, although we could readily detect donor-derived IL-21+CD4+ T cells infiltrating the tumor, it remains unclear whether CD4+ T-cell-mediated CX3CR1+CD8+ T cell differentiation can occur directly within the immunosuppressive tumor microenvironment or in secondary lymphoid tissues (where the Ly108+ progenitor population preferentially resides). A determination of whether CD4+ T cell help is essential in particular anatomical locations for driving the formation of CX3CR1+ CD8+ T cells will have important implications for optimizing immunotherapy. Notably, our data identify that the differentiation defect of unhelped CD8+ T cells was associated with diminished T-bet and upregulated Eomes expression. This finding is consistent with the Cx3cr1+ cell subset displaying uniformly high expression of Tbx21, whereas Pdcd1+ exhausted cells exhibited low Tbx21 expression but high Eomes expression (Figures S2E and S2F). However, whether differential expression of these two transcription factors is responsible for the differentiation trajectory of virus-specific CD8+ T cells along a PD-1hi exhausted or CX3CR1+ developmental pathway remains to be further investigated. Moreover, although it is well appreciated that targeting the PD-1-PD-L1 axis can re-invigorate T cell responses during
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
persistent infection and cancer (Barber et al., 2006; Wherry and Kurachi, 2015), our data indicate that PD-L1 blockade is insufficient to rescue the defect in CX3CR1+ CD8+ T cell differentiation that occurs in un-helped CD8+ T cells. These findings may be of particular relevance when considering immunotherapy to treat diseases wherein low CD4+ T cell numbers are often a common occurrence, such as during HIV infection (Klatzmann et al., 1984; Masur et al., 1989). Additionally, although several recent studies have identified that the TCF-1hi progenitor population is the subset that provides a proliferative burst in response to PD-L1 blockade (Leong et al., 2016; He et al., 2016; Im et al., 2016; Utzschneider et al., 2016), we speculate that maintaining a pool of cytotoxic CX3CR1+ CD8+ T cells in conjunction with the TCF-1hi (Slamf6+) progenitor population is likely required for optimizing the therapeutic efficacy of blocking the PD-1-PD-L1 inhibitory axis. Taken together, our study uncovers broad transcriptional heterogeneity among CD8+ T cells responding to persistent infection and supports a previously unrecognized paradigm of CD8+ T cell differentiation during chronic viral infection. Moreover, our data further indicate that CD4+ T cell help in the form of IL21 may potentially be harnessed to bolster the formation of protective cytolytic CX3CR1+ CD8+ T cells and improve control over chronic viral infection and tumor progression. STAR+METHODS Detailed methods are provided in the online version of this paper and include the following: d d d
d
d d
KEY RESOURCES TABLE LEAD CONTACT AND MATERIALS AVAILABILITY EXPERIMENTAL MODEL AND SUBJECT DETAILS B Mice and LCMV Cl13 infection B Tumor cell lines and Tumor Induction METHOD DETAILS + B Generation of Polyclonal Tumor Reactive CD4 T Cells B Immune Cell isolation from solid tumors B Flow cytometry B IncuCyte cytotoxicity assay B Administration of biologics B Cell transfer experiments B Mixed bone marrow (MBM) chimeras B Single-cell RNA sequencing B RNA isolation and quantitative real-time PCR QUANTIFICATION AND STATISTICAL ANALYSIS DATA AND CODE AVAILABILITY
SUPPLEMENTAL INFORMATION Supplemental Information can be found online at https://doi.org/10.1016/j. immuni.2019.10.009.
T32-GM080202. This research was completed in part with computational resources and technical support provided by the Research Computing Center at MCW.
AUTHOR CONTRIBUTIONS R.Z., D.S., G.X., C.N., X.W., and W.C. designed and performed the experiments and analyzed the data. D.S. analyzed RNA-seq datasets. A.Z. provided helpful insight and contributed with key reagents, including IL-21/ P14 cells and bone marrow from IL-21/ mice. R.Z., D.S., and W.C. wrote the manuscript. W.C. supervised the study.
DECLARATION OF INTERESTS The authors declare no competing interests. Received: October 30, 2018 Revised: August 1, 2019 Accepted: October 22, 2019 Published: December 17, 2019 REFERENCES Ahmadzadeh, M., Johnson, L.A., Heemskerk, B., Wunderlich, J.R., Dudley, M.E., White, D.E., and Rosenberg, S.A. (2009). Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 114, 1537–1544. Barber, D.L., Wherry, E.J., Masopust, D., Zhu, B., Allison, J.P., Sharpe, A.H., Freeman, G.J., and Ahmed, R. (2006). Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439, 682–687. Battegay, M., Moskophidis, D., Rahemtulla, A., Hengartner, H., Mak, T.W., and Zinkernagel, R.M. (1994). Enhanced establishment of a virus carrier state in adult CD4+ T-cell-deficient mice. J. Virol. 68, 4700–4704. Blackburn, S.D., Shin, H., Haining, W.N., Zou, T., Workman, C.J., Polley, A., Betts, M.R., Freeman, G.J., Vignali, D.A., and Wherry, E.J. (2009). Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat. Immunol. 10, 29–37. Dominguez, C.X., Amezquita, R.A., Guan, T., Marshall, H.D., Joshi, N.S., Kleinstein, S.H., and Kaech, S.M. (2015). The transcription factors ZEB2 and T-bet cooperate to program cytotoxic T cell terminal differentiation in response to LCMV viral infection. J. Exp. Med. 212, 2041–2056. Elsaesser, H., Sauer, K., and Brooks, D.G. (2009). IL-21 is required to control chronic viral infection. Science 324, 1569–1572. Fro¨hlich, A., Kisielow, J., Schmitz, I., Freigang, S., Shamshiev, A.T., Weber, J., Marsland, B.J., Oxenius, A., and Kopf, M. (2009). IL-21R on T cells is critical for sustained functionality and control of chronic viral infection. Science 324, 1576–1580. Gattinoni, L., Finkelstein, S.E., Klebanoff, C.A., Antony, P.A., Palmer, D.C., Spiess, P.J., Hwang, L.N., Yu, Z., Wrzesinski, C., Heimann, D.M., et al. (2005). Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J. Exp. Med. 202, 907–912. Hanada, K.I., Yu, Z., Chappell, G.R., Park, A.S., and Restifo, N.P. (2019). An effective mouse model for adoptive cancer immunotherapy targeting neoantigens. JCI Insight 4, 124405.
ACKNOWLEDGMENTS
He, R., Hou, S., Liu, C., Zhang, A., Bai, Q., Han, M., Yang, Y., Wei, G., Shen, T., Yang, X., et al. (2016). Follicular CXCR5- expressing CD8(+) T cells curtail chronic viral infection. Nature 537, 412–428.
This work is supported by NIH grants AI125741 (W.C.) and DK108557 (D.S.) and by American Cancer Society (ACS) Research Scholar Grant and A Healthier Wisconsin (AHW) Grant (W.C.). R.Z. is supported by the Cancer Research Institute Irvington Fellowship. G.X. is supported by The Elizabeth Elser Doolittle Postdoctoral Fellowship. D.S. is a member of the Medical Scientist Training Program at MCW, which is partially supported by a training grant from NIGMS
Hudson, W.H., Gensheimer, J., Hashimoto, M., Wieland, A., Valanparambil, R.M., Li, P., Lin, J.-X., Konieczny, B.T., Im, S.J., Freeman, G.J., et al. (2019). Proliferating Transitory T Cells with an Effector-like Transcriptional Signature Emerge from PD-1+ Stem-like CD8+ T Cells during Chronic Infection. Immunity 51. Published online November 26, 2019. https://doi.org/10.1016/j. immuni.2019.11.002.
Immunity 53, 1–15, December 15, 2020 13
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
Im, S.J., Hashimoto, M., Gerner, M.Y., Lee, J., Kissick, H.T., Burger, M.C., Shan, Q., Hale, J.S., Lee, J., Nasti, T.H., et al. (2016). Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421. Jin, X., Bauer, D.E., Tuttleton, S.E., Lewin, S., Gettie, A., Blanchard, J., Irwin, C.E., Safrit, J.T., Mittler, J., Weinberger, L., et al. (1999). Dramatic rise in plasma viremia after CD8(+) T cell depletion in simian immunodeficiency virus-infected macaques. J. Exp. Med. 189, 991–998. Kaech, S.M., and Cui, W. (2012). Transcriptional control of effector and memory CD8+ T cell differentiation. Nat. Rev. Immunol. 12, 749–761. Kahan, S.M., Wherry, E.J., and Zajac, A.J. (2015). T cell exhaustion during persistent viral infections. Virology 479-480, 180–193. Klatzmann, D., Barre´-Sinoussi, F., Nugeyre, M.T., Danquet, C., Vilmer, E., Griscelli, C., Brun-Veziret, F., Rouzioux, C., Gluckman, J.C., Chermann, J.C., et al. (1984). Selective tropism of lymphadenopathy associated virus (LAV) for helper-inducer T lymphocytes. Science 225, 59–63. Klebanoff, C.A., Gattinoni, L., Torabi-Parizi, P., Kerstann, K., Cardones, A.R., Finkelstein, S.E., Palmer, D.C., Antony, P.A., Hwang, S.T., Rosenberg, S.A., et al. (2005). Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc. Natl. Acad. Sci. USA 102, 9571–9576. Kurtulus, S., Madi, A., Escobar, G., Klapholz, M., Nyman, J., Christian, E., Pawlak, M., Dionne, D., Xia, J., Rozenblatt-Rosen, O., et al. (2019). Checkpoint blockade immunotherapy induces dynamic changes in PD1–CD8+ tumor-infiltrating T cells. Immunity 50, 181–194.e6. Leong, Y.A., Chen, Y., Ong, H.S., Wu, D., Man, K., Deleage, C., Minnich, M., Meckiff, B.J., Wei, Y., Hou, Z., et al. (2016). CXCR5(+) follicular cytotoxic T cells control viral infection in B cell follicles. Nat. Immunol. 17, 1187–1196. Liang, X., Fu, C., Cui, W., Ober-Blo¨baum, J.L., Zahner, S.P., Shrikant, P.A., Clausen, B.E., Flavell, R.A., Mellman, I., and Jiang, A. (2014). b-catenin mediates tumor-induced immunosuppression by inhibiting cross-priming of CD8+ T cells. J. Leukoc. Biol. 95, 179–190.
Paley, M.A., Kroy, D.C., Odorizzi, P.M., Johnnidis, J.B., Dolfi, D.V., Barnett, B.E., Bikoff, E.K., Robertson, E.J., Lauer, G.M., Reiner, S.L., and Wherry, E.J. (2012). Progenitor and terminal subsets of CD8+ T cells cooperate to contain chronic viral infection. Science 338, 1220–1225. Papalexi, E., and Satija, R. (2018). Single-cell RNA sequencing to explore immune cell heterogeneity. Nat. Rev. Immunol. 18, 35–45. €rki, K., Hengartner, H., and Pircher, H., Moskophidis, D., Rohrer, U., Bu Zinkernagel, R.M. (1990). Viral escape by selection of cytotoxic T cell-resistant virus variants in vivo. Nature 346, 629–633. Purwar, R., Schlapbach, C., Xiao, S., Kang, H.S., Elyaman, W., Jiang, X., Jetten, A.M., Khoury, S.J., Fuhlbrigge, R.C., Kuchroo, V.K., et al. (2012). Robust tumor immunity to melanoma mediated by interleukin-9-producing T cells. Nat. Med. 18, 1248–1253. Qiu, X., Mao, Q., Tang, Y., Wang, L., Chawla, R., Pliner, H.A., and Trapnell, C. (2017). Reversed graph embedding resolves complex single-cell trajectories. Nat. Methods 14, 979–982. Sakuishi, K., Apetoh, L., Sullivan, J.M., Blazar, B.R., Kuchroo, V.K., and Anderson, A.C. (2010). Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J. Exp. Med. 207, 2187–2194. Satija, R., Farrell, J.A., Gennert, D., Schier, A.F., and Regev, A. (2015). Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502. Schmitz, J.E., Kuroda, M.J., Santra, S., Sasseville, V.G., Simon, M.A., Lifton, M.A., Racz, P., Tenner-Racz, K., Dalesandro, M., Scallon, B.J., et al. (1999). Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes. Science 283, 857–860. Shulman, Z., Gitlin, A.D., Weinstein, J.S., Lainez, B., Esplugues, E., Flavell, R.A., Craft, J.E., and Nussenzweig, M.C. (2014). Dynamic signaling by T follicular helper cells during germinal center B cell selection. Science 345, 1058–1062.
Lu, Y., Hong, B., Li, H., Zheng, Y., Zhang, M., Wang, S., Qian, J., and Yi, Q. (2014). Tumor-specific IL-9-producing CD8+ Tc9 cells are superior effector than type-I cytotoxic Tc1 cells for adoptive immunotherapy of cancers. Proc. Natl. Acad. Sci. USA 111, 2265–2270.
Siddiqui, I., Schaeuble, K., Chennupati, V., Fuertes Marraco, S.A., CalderonCopete, S., Pais Ferreira, D., Carmona, S.J., Scarpellino, L., Gfeller, D., Pradervand, S., et al. (2019). Intratumoral Tcf1+PD-1+CD8+ T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 50, 195–211.e10.
Masur, H., Ognibene, F.P., Yarchoan, R., Shelhamer, J.H., Baird, B.F., Travis, W., Suffredini, A.F., Deyton, L., Kovacs, J.A., Falloon, J., et al. (1989). CD4 counts as predictors of opportunistic pneumonias in human immunodeficiency virus (HIV) infection. Ann. Intern. Med. 111, 223–231.
Søndergaard, H., Galsgaard, E.D., Bartholomaeussen, M., Straten, P.T., Odum, N., and Skak, K. (2010). Intratumoral interleukin-21 increases antitumor immunity, tumor-infiltrating CD8+ T-cell density and activity, and enlarges draining lymph nodes. J. Immunother. 33, 236–249.
Matloubian, M., Concepcion, R.J., and Ahmed, R. (1994). CD4+ T cells are required to sustain CD8+ cytotoxic T-cell responses during chronic viral infection. J. Virol. 68, 8056–8063.
Tian, Y., Cox, M.A., Kahan, S.M., Ingram, J.T., Bakshi, R.K., and Zajac, A.J. (2016). A context-dependent role for IL-21 in modulating the differentiation, distribution, and abundance of effector and memory CD8 T cell subsets. J. Immunol. 196, 2153–2166.
McCausland, M.M., and Crotty, S. (2008). Quantitative PCR technique for detecting lymphocytic choriomeningitis virus in vivo. J. Virol. Methods 147, 167–176. Moroz, A., Eppolito, C., Li, Q., Tao, J., Clegg, C.H., and Shrikant, P.A. (2004). IL-21 enhances and sustains CD8+ T cell responses to achieve durable tumor immunity: comparative evaluation of IL-2, IL-15, and IL-21. J. Immunol. 173, 900–909. Muranski, P., Boni, A., Antony, P.A., Cassard, L., Irvine, K.R., Kaiser, A., Paulos, C.M., Palmer, D.C., Touloukian, C.E., Ptak, K., et al. (2008). Tumorspecific Th17-polarized cells eradicate large established melanoma. Blood 112, 362–373. Omilusik, K.D., Best, J.A., Yu, B., Goossens, S., Weidemann, A., Nguyen, J.V., Seuntjens, E., Stryjewska, A., Zweier, C., Roychoudhuri, R., et al. (2015). Transcriptional repressor ZEB2 promotes terminal differentiation of CD8+ effector and memory T cell populations during infection. J. Exp. Med. 212, 2027–2039. Overwijk, W.W., and Restifo, N.P. (2001). B16 as a mouse model for human melanoma. Curr. Protoc. Immunol. Chapter 20, Unit 20.1. Overwijk, W.W., Theoret, M.R., Finkelstein, S.E., Surman, D.R., de Jong, L.A., Vyth-Dreese, F.A., Dellemijn, T.A., Antony, P.A., Spiess, P.J., Palmer, D.C., et al. (2003). Tumor regression and autoimmunity after reversal of a functionally tolerant state of self-reactive CD8+ T cells. J. Exp. Med. 198, 569–580.
14 Immunity 53, 1–15, December 15, 2020
Trapnell, C., Cacchiarelli, D., Grimsby, J., Pokharel, P., Li, S., Morse, M., Lennon, N.J., Livak, K.J., Mikkelsen, T.S., and Rinn, J.L. (2014). The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386. Utzschneider, D.T., Charmoy, M., Chennupati, V., Pousse, L., Ferreira, D.P., Calderon-Copete, S., Danilo, M., Alfei, F., Hofmann, M., Wieland, D., et al. (2016). T cell factor 1-expressing memory-like CD8(+) T cells sustain the immune response to chronic viral infections. Immunity 45, 415–427. Ve´gran, F., Berger, H., Boidot, R., Mignot, G., Bruchard, M., Dosset, M., Chalmin, F., Re´be´, C., De´range`re, V., Ryffel, B., et al. (2014). The transcription factor IRF1 dictates the IL-21-dependent anticancer functions of TH9 cells. Nat. Immunol. 15, 758–766. Villani, A.C., Satija, R., Reynolds, G., Sarkizova, S., Shekhar, K., Fletcher, J., Griesbeck, M., Butler, A., Zheng, S., Lazo, S., et al. (2017). Single-cell RNAseq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science 356, eaah4573. Waltman, L., and Jan van Eck, N. (2013). A smart local moving algorithm for large-scale modularity-based community detection. Eur. Phys. J. B 86, 471. Wang, J., Saffold, S., Cao, X., Krauss, J., and Chen, W. (1998). Eliciting T cell immunity against poorly immunogenic tumors by immunization with dendritic cell-tumor fusion vaccines. J. Immunol. 161, 5516–5524.
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
Wherry, E.J., and Kurachi, M. (2015). Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 15, 486–499.
Yi, J.S., Du, M., and Zajac, A.J. (2009a). A vital role for interleukin-21 in the control of a chronic viral infection. Science 324, 1572–1576.
Wherry, E.J., Blattman, J.N., Murali-Krishna, K., van der Most, R., and Ahmed, R. (2003). Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment. J. Virol. 77, 4911–4927.
Zajac, A.J., Blattman, J.N., Murali-Krishna, K., Sourdive, D.J., Suresh, M., Altman, J.D., and Ahmed, R. (1998). Viral immune evasion due to persistence of activated T cells without effector function. J. Exp. Med. 188, 2205–2213.
Xin, G., Schauder, D.M., Lainez, B., Weinstein, J.S., Dai, Z., Chen, Y., Esplugues, E., Wen, R., Wang, D., Parish, I.A., et al. (2015). A critical role of IL-21-induced BATF in sustaining CD8-T-cell-mediated chronic viral control. Cell Rep. 13, 1118–1124.
Zhang, D.J., Wang, Q., Wei, J., Baimukanova, G., Buchholz, F., Stewart, A.F., Mao, X., and Killeen, N. (2005). Selective expression of the Cre recombinase in late-stage thymocytes using the distal promoter of the Lck gene. J. Immunol. 174, 6725–6731.
Immunity 53, 1–15, December 15, 2020 15
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
STAR+METHODS KEY RESOURCES TABLE
REAGENT or RESOURCE
SOURCE
IDENTIFIER
Mouse strains C57BL/6
Charles River
N/A
Cx3cr1DTR mice
The Jackson Lab
Stock # 025629
dLck-cre-recombinase mice
The Jackson Lab
Stock # 012837
IL-21-tRFP reporter mice
(Shulman et al., 2014)
N/A
Anti-mouse PD.1 (RMP1-30)
Biolegend
Cat #109110; RRID:AB_572017
Anti-mouse Thy1.1 (OX-7)
Biolegend
Cat #202506; RRID:AB_492882
Anti-mouse CXCR3 (CXCR3-173)
Biolegend
Cat #126512; RRID:AB_1088993
Anti-mouse CXCR5 (L138D7)
Biolegend
Cat #145526; RRID:AB_2566799
Anti-mouse CXCR6 (SA051D1)
Biolegend
Cat #151117; RRID:AB_2721700
Anti-mouse CX3CR1 (SA011F11)
Biolegend
Cat #149014; RRID:AB_2565698
Anti-mouse Ly108 (330-AJ)
Biolegend
Cat #134608; RRID:AB_2188093
Anti-mouse Tim3
BD Biosciences
Cat #747625; RRID:AB_2744191
Anti-mouse 2B4 (M2B4 (B6)458.1)
Biolegend
Cat #133503; RRID:AB_1595624
Anti-mouse KLRG1 (2F1)
Biolegend
Cat #138416; RRID:AB_2561736
Anti-mouse KLRa9 (Ly-49I; YLI-90)
eBioscience
Cat #12-5895-82; RRID:AB_466021
Anti-mouse CD127 (A7R34)
Biolegend
Cat #135014; RRID:AB_1937265
Flow Cytometry Reagents
Anti-mouse CD107a (1D4B)
Biolegend
Cat#121616; RRID:AB_10643268
Anti-mouse TNF-a (MP6-XT22)
Biolegend
Cat#506306; RRID:AB_315427
Anti-mouse IFN-g (XMG1.2)
Biolegend
Cat#505826; RRID:AB_2295770
Anti-human/mouse Granzyme B (GB11)
Invitrogen
Cat#GRB04; RRID:AB_2536538
Anti-TCF-1 (C63D9)
Cell Signaling
Cat#2203S; RRID:AB_2199302
Anti-mouse BATF (D7C5)
Cell Signaling
Cat#8638S; RRID:AB_11141425
Anti-mouse Eomes (Dan11mag)
Invitrogen
Cat#25-4875-82; RRID:AB_2573454
Anti-mouse T-bet (4B10)
Biolegend
Cat#644806; RRID:AB_1595488
LCMV DbGP33 tetramer
Made in house
N/A
Brefeldin A Solution (1,000X)
Biolegend
Cat#420601
Fixation Buffer
Biolegend
Cat#420801
True Nuclear Transcription Factor Buffer Set
Biolegend
Cat#424401
Rafi Ahmed
Grew up in house
ATCC
Cat# CRL-6475
RNAlater-ICE
Invitrogen
Cat#AM7030
KAVYNFATM (GP33-41) peptide
GenScript
RP20257
GP-F: CATTCACCTGGACTTTGTCAGACTC
(McCausland and Crotty, 2008) IDT
N/A
GP-R: GCAACTGCTGTGTTCCCGAAAC
(McCausland and Crotty, 2008) IDT
N/A
Experimental Models: LCMV LCMV Clone (Cl13) virus strain Experimental Models: Tumor Cell Lines B16/F10 tumor cell line Chemicals, Peptides and Recombinant Proteins
Oligonucleotides
Critical Commercial Assays Incucyte Annexin V Red Reagent
Essen Bioscience
Cat#4641
QiAmp MinElute Virus Spin Kit
QIAGEN
Cat#57704 (Continued on next page)
e1 Immunity 53, 1–15.e1–e4, December 15, 2020
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
Continued REAGENT or RESOURCE
SOURCE
IDENTIFIER
RNAqueous-Micro Kit
Thermofisher
Cat#AM1931
Mouse Treatment reagents Anti-mouse CD4 Mab (GK1.5)
Bioxcell
Cat#BE0003-1
Anti-PD-L1 (10F.9G2)
Bioxcell
Cat#BE0101
Diptheria Toxin
Sigma-Aldrich
Cat#D0564
10X Genomics
Cat#PN-120267
Single cell RNA sequencing Chromium Single Cell 30 Library & Gel Bead Kit v2 Chromium Single Cell A Chip Kit
10X Genomics
Cat#PN-1000009
Chromium i7 Multiplex Kit
10X Genomics
Cat# PN-120262
Dynabeads MyOne Silane
Thermofisher
Cat#37002D
SPRIselect Reagent Kit
Beckman Coulter
Cat#B23318
Kappa NGS quantification kit
KAPABiosystems
Cat#KK4824
NextSeq 500/550 High Output Kit v2.5 (150 cycles)
Illumina
Cat#20024907
Cell Ranger
10X Genomics
https://support.10xgenomics.com/singlecell-gene-expression/software/pipelines/ latest/installation
Seurat
(Satija et al., 2015)
https://satijalab.org/seurat/
Monocle-2
(Qiu et al., 2017) (Trapnell et al., 2014)
http://cole-trapnell-lab.github.io/monoclerelease/docs/
In this paper
GSE 129139
Flowjo Version 10.5.3
Tree Star
N/A
Prism 8
Graphpad Software
N/A
Computational analysis
Datasets Day 8 and Day 30 single cell RNA sequencing on DbGP33+ CD8 T cells Statistical or Data Analysis
LEAD CONTACT AND MATERIALS AVAILABILITY Further information and requests for resources should be directed to and will be fulfilled by the Lead Contact, Weiguo Cui (weiguo.
[email protected]). EXPERIMENTAL MODEL AND SUBJECT DETAILS Mice and LCMV Cl13 infection Six to eight-week-old female C57BL/6 and CD45.1 congenic mice were obtained through the National Cancer Institute grantees program (Frederick, MD). IL-21-tRFP mice were obtained from Dr. Joseph Craft, Yale School of Medicine, CT. Mice were bred and maintained in a closed breeding facility, and mouse handling conformed to the requirements of the Institutional Animal Care and Use Guidelines of Medical College of Wisconsin. Cx3cr1DTR mice (stock #:025629) and dLck-cre-recombinase mice (stock #:012837) were purchased from Jackson and crossed to one another to generate either Cx3cr1DTRdLck-cre– or Cx3cr1DTRdLckcre+ mice. Mice were infected with LCMV clone 13 (Cl13), which was intravenously injected into mice (2 3 106 PFU/mouse) to establish chronic infection. LCMV Cl13 was prepared by a single passage on BHK21 cells and viral titers were determined by plaque formation assay on Vero cells. Tumor cell lines and Tumor Induction B16-F10 cells were obtained from ATCC and cultured in high-glucose DMEM (Cellgro) supplemented with 10% (vol/vol) FBS. Melanoma tumors were established by injecting 2 3 105 B16-F10 cells s.c. on one flank of the C57BL/6 mice. Tumor growth was monitored by measuring with calipers every other day, and tumor volume was calculated as length * (width)2/2
Immunity 53, 1–15.e1–e4, December 15, 2020 e2
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
METHOD DETAILS Generation of Polyclonal Tumor Reactive CD4+ T Cells Bone marrow cells were isolated from C57BL/6 mice and cultured in RPMI (Cellgro) medium with 10% (vol/vol) FBS and 200 ng/mL Flt3L for 1 wk. On day 7, DCs were harvested and incubated with freeze-thawed tumor lysates at a ratio of one tumor cell equivalent to one DC (i.e., 1:1) as described (Liang et al., 2014). After 18 hours of incubation, DCs were harvested and maturated with LPS for 4 hours. The mature DCs and purified IL-21-tRFP CD4+ T cells were mixed in 1:2 ratio and cultured under Th17 cell conditions using IL-6 (50 ng/mL), TGF-b (10ng/mL), IL-23 (20ng/mL), and IL-21 (10ng/mL) and also with anti-IL-4 and anti-IFN-g antibodies (10ug/mL). Then, activated CD4 T cells were sort-purified based on IL-21-tRFP expression and adoptively transferred into recipient mice that were inoculated with B16-F10 7-10 days prior. Immune Cell isolation from solid tumors Dissected tumor tissues were cut into small pieces and digested with 0.7 mg/mL collagenase XI Sigma-Aldrich) and 30 mg/mL type IV bovine pancreatic DNase (Sigma-Aldrich) for 45 min at 37 C. Immune cells were then isolated by centrifugation with Lymphocyte Cell Separation Medium (Cedarlane Labs) Flow cytometry All flow cytometry data were acquired on an LSRII (BD Biosciences, CA) and analyzed by FlowJo (Treestar, OR). Lymphocytes were isolated from tissues including spleen, blood, liver, bone marrow, and inguinal lymph nodes as described previously. Cells were then stained with GP33-41 tetramer and antibodies against cell surface antigens for 30-60 minutes at 4 degrees. Transcription factor staining was performed using the True Nuclear transcription factor buffer set (Biolegend). BrdU was administered to mice i.p. and measured in LCMV-specific cells according to the manufactures protocol (BD Bioscience). IncuCyte cytotoxicity assay Target EL4 cells were pulsed with either GP33 or irrelevant ovalbumin SIINFEKL (OT-I) peptide for 2 hours and then incubated with sorted GP33-41+ CD8+ T cells (5:1 E: T) in the presence of IncuCyte Annexin V Red Reagent (Essen BioScience Cat #4641), which enables real-time detection of cells undergoing apoptosis. Images were taken every 2 hours and analyzed by incuCyte S3 LiveCell Analysis System. Administration of biologics To deplete CD4 T cells, mice received i.p. injections of 500 mg anti-CD4 antibody (clone GK1.5 from BioXCell, NH) one day before LCMV Cl13 infection. InVivoMab anti-mouse PD-L1 (clone 10F.9G2) was purchased from BioXCell and administered in 200 ug doses on days 16, 19, and 22 p.i. Cell transfer experiments For recall experiments, CD8+CD44hi CX3CR1+, Ly108+ or CX3CR1-Ly108- subsets were sorted from LCMV-Cl13-infected mice on day 45 p.i. using an Aria IIIu cell sorter. Next we adaptively transferred 60k cells of each respective subset into separate groups of congenically marked naive Ly5.1 mice that were subsequently challenged with LCMV-Cl13 one day later. In another experiment, 80k cells of each respective subset were transferred in experimental mice and identical results were observed. For P14 transfer experiments, congenically marked WT or Il21r/ P14 cells were co-transferred at a 1:1 ratio (5000 cells each) into naive Ly5.1 mice that were then infected one day later. Mixed bone marrow (MBM) chimeras For MBM chimera experiments, recipient mice were irradiated with 6.5 and 5.5 Gy separated by 8 h. Bone marrow from various donor mice (as depicted in Figures) were mixed at the indicated ratios, and a total of 6 3 106 cells were transferred i.v. Mice were maintained on oral sulfamethoxazole for 2 weeks. Experimental MBM chimera mice were either infected with LCMV Cl13, or injected with B16-F10 melanoma cells at 8 weeks post reconstitution. Single-cell RNA sequencing LCMV-specific CD8 T cells were FACS-sorted from LCMV Cl13-infected mice on days 8 and 30 p.i. and were loaded on the Chromium Controller (10x Genomics). Single-cell RNA-seq libraries were prepared using the Chromium Single Cell 30 v2 Reagent Kit (10x Genomics) according to manufacturer’s protocol. Libraries were loaded onto an Illumina NextSeq with the NextSeq 500/550 High Output Kit v2 (150 cycles) (FC-404-2002, Illumina) with the following conditions: 26 cycles for read 1, 98 cycles for read 2, and 8 cycles for i7 index. Python Run Downloader (Illumina) was used to download raw sequencing data. Cell Ranger (10x Genomics) functions mkfastq and count were used to demultiplex the sequencing data and generate gene-barcode matrices, respectively. All scRNA-seq analyses were performed in R (version 3.4.0) using the package Seurat (version 2.2.0) (Satija et al., 2015) and Monocle (Trapnell et al., 2014). Number of genes detected per cell, number of UMIs, and percent mitochondrial genes were plotted, and outliers were removed (number of genes over 2,500, number of UMIs over 8,000, and percent mitochondrial genes over 0.08) to filter out doublets
e3 Immunity 53, 1–15.e1–e4, December 15, 2020
Please cite this article in press as: Zander et al., CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer, Immunity (2019), https://doi.org/10.1016/j.immuni.2019.10.009
and dead cells. Principal component analysis was performed, and the top 9 most statistically significant principal components were used for t-SNE analysis, with 2,000 iterations and a perplexity parameter of 30. RNA isolation and quantitative real-time PCR Fractions of spleens were snap-frozen on dry ice and subsequently stored in a 80 C freezer. Samples were stabilized in RNAlaterICE (ThermoFisher) before proceeding with tissue homogenization and RNA isolation using the RNAqueous-Micro Kit (ThermoFisher) according to the manufacturer’s instructions. For sera samples, RNA was extracted from 10ul of sera using QiAamp MinElute Virus Spin Kit (QIAGEN). Determination of viral load by qPCR was performed as has been previously described (McCausland and Crotty, 2008) and the following primers were used during the qPCR reaction: GP-R (S pos. 970-991), GCAACTGCTGTGTTCCCGAAAC. GPF (S pos. 877-901), CATTCACCTGGACTTTGTCAGACTC. Amplification was done for 40 cycles with each cycle consisting of two steps: 95 C, 15 s; and 60 C, 30 s. Standard curves were generated using serial dilutions of a gene fragment (gblocks IDT) derived from Lymphocytic choriomeningitis virus clone 13 segment S, with the following sequence: 50 - AGA GAA GAC TAA GTT CCT CAC TAG GAG ACT AGC GGG CAC ATT CAC CTG GAC TTT GTC AGA CTC TTC AGG GGT GGA GAA TCC AGG TGG TTA TTG CCT GAC CAA ATG GAT GAT TCT TGC TGC AGA GCT TAA GTG TTT CGG GAA CAC AGC AGT TGC GAA ATG CAA TGT AAA TCA TGA TGA AGA ATT CTG TG 30 . QUANTIFICATION AND STATISTICAL ANALYSIS Statistical tests were performed using Graphpad Prism 7. P values were calculated using either two-tailed unpaired Student’s t tests or one-way ANOVA while correcting for multiple comparisons via the Tukey method. DATA AND CODE AVAILABILITY The single-cell RNA-seq data from this paper is available in the GEO database with the accession code GSE129139. All other raw data are available from the Lead Contact upon request.
Immunity 53, 1–15.e1–e4, December 15, 2020 e4