interleukin-10 directly inhibits cd8+ t cell function by ... · interleukin-10 directly inhibits...

20
Article Interleukin-10 Directly Inhibits CD8 + T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity Graphical Abstract Highlights d IL-10 directly increases the threshold for CD8 + T cell activation d Induction of Mgat5 is necessary for IL-10-mediated repression d The galectin-glycoprotein surface lattice promotes T cell dysfunction d Disruption of galectin binding restores T cell function and viral control Authors Logan K. Smith, Giselle M. Boukhaled, Stephanie A. Condotta, ..., Naglaa H. Shoukry, Connie M. Krawczyk, Martin J. Richer Correspondence [email protected] In Brief The mechanisms by which IL-10 facilitates the establishment of chronic infections are not fully understood. Smith et al. demonstrate that during chronic infections, IL-10 upregulates N-glycan branching on CD8 + T cell surface glycoproteins, which reduces signal transduction downstream of the T cell receptor and decreases CD8 + T cell antigen sensitivity and capacity to control pathogen burden. Smith et al., 2018, Immunity 48, 1–14 February 20, 2018 ª 2018 Elsevier Inc. https://doi.org/10.1016/j.immuni.2018.01.006

Upload: others

Post on 26-Jun-2020

34 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

Article

Interleukin-10 Directly Inh

ibits CD8+ T Cell Functionby Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity

Graphical Abstract

Highlights

d IL-10 directly increases the threshold for CD8+ T cell

activation

d Induction of Mgat5 is necessary for IL-10-mediated

repression

d The galectin-glycoprotein surface lattice promotes T cell

dysfunction

d Disruption of galectin binding restores T cell function and viral

control

Smith et al., 2018, Immunity 48, 1–14February 20, 2018 ª 2018 Elsevier Inc.https://doi.org/10.1016/j.immuni.2018.01.006

Authors

Logan K. Smith, Giselle M. Boukhaled,

Stephanie A. Condotta, ...,

Naglaa H. Shoukry,

Connie M. Krawczyk, Martin J. Richer

[email protected]

In Brief

The mechanisms by which IL-10

facilitates the establishment of chronic

infections are not fully understood. Smith

et al. demonstrate that during chronic

infections, IL-10 upregulates N-glycan

branching on CD8+ T cell surface

glycoproteins, which reduces signal

transduction downstream of the T cell

receptor and decreases CD8+ T cell

antigen sensitivity and capacity to control

pathogen burden.

Page 2: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

Immunity

Article

Interleukin-10 Directly Inhibits CD8+ T CellFunction by Enhancing N-Glycan Branchingto Decrease Antigen SensitivityLogan K. Smith,1,2 Giselle M. Boukhaled,3 Stephanie A. Condotta,1,2 Sabrina Mazouz,4,5 Jenna J. Guthmiller,6

Rahul Vijay,7 Noah S. Butler,6,7 Julie Bruneau,4,8 Naglaa H. Shoukry,4,9 Connie M. Krawczyk,1,3 and Martin J. Richer1,2,10,*1Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada2Microbiome and Disease Tolerance Centre, McGill University, Montreal, QC, Canada3Department of Physiology, Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada4Centre de Recherche du Centre hospitalier de l’Universite de Montreal (CRCHUM), Montreal, QC, Canada5Department of Microbiology, Immunology and Infectiology, Universite de Montreal, Montreal, QC, Canada6Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA7Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA8Department of Family Medicine and Emergency Medicine, Universite de Montreal, Montreal, QC, Canada9Department of Medicine, Universite de Montreal, Montreal, QC, Canada10Lead Contact*Correspondence: [email protected]

https://doi.org/10.1016/j.immuni.2018.01.006

SUMMARY

Chronic viral infections remain a global healthconcern. The early events that facilitate viral persis-tence have been linked to the activity of the immuno-regulatory cytokine IL-10. However, the mechanismsby which IL-10 facilitates the establishment ofchronic infection are not fully understood. Herein,we demonstrated that the antigen sensitivity ofCD8+ T cells was decreased during chronic infectionand that this was directly mediated by IL-10.Mechanistically, we showed that IL-10 inducedthe expression of Mgat5, a glycosyltransferase thatenhances N-glycan branching on surface glyco-proteins. Increased N-glycan branching on CD8+

T cells promoted the formation of a galectin 3-medi-ated membrane lattice, which restricted the interac-tion of key glycoproteins, ultimately increasing theantigenic threshold required for T cell activation.Our study identified a regulatory loop in which IL-10directly restricts CD8+ T cell activation and functionthrough modification of cell surface glycosylationallowing the establishment of chronic infection.

INTRODUCTION

Chronic viral infections, such as human immunodeficiency virus

(HIV), hepatitis C virus (HCV), and hepatitis B virus (HBV),

remain a tremendous global health burden threatening a

significant proportion of the population. These infections lead

to progressive immune dysfunction, including the functional

exhaustion and eventual deletion of the responding T lympho-

cytes (Wherry and Kurachi, 2015). While the consequences of

chronic viral infections on the host immune system are well

described, less is known of the early events that allow for the

establishment of viral persistence. CD8+ T cells play a critical

role in the immune response to viral infections and are central

to the capacity of the host to prevent the establishment of

chronic infection (Barber et al., 2006). Efficient pathogen con-

trol relies on the capacity of CD8+ T cells to rapidly respond

and develop effector functions in the presence of low levels

of antigen, also known as antigen sensitivity or functional avid-

ity (Alexander-Miller, 2005; Vigano et al., 2012; Walker et al.,

2010). The importance of antigen sensitivity is best illustrated

by the findings that patients who can resolve multiple HCV

infections harbor highly sensitive HCV-specific CD8+ T cells

compared to patients who progress to chronic infection

(Abdel-Hakeem et al., 2017). Thus, antigen sensitivity of the re-

sponding CD8+ T cells may be a key characteristic that deter-

mines whether the immune response is able to rapidly and

efficiently control the invading pathogen.

The affinity of the T cell receptor (TCR) for peptide antigen is

relatively low and, as opposed to the B cell receptor, remains

fixed during the lifetime of the cell. However, the antigen sensi-

tivity of CD8+ T cells is modulated during infection at both the

population (Busch and Pamer, 1999; Malherbe et al., 2004;

Zehn et al., 2009) and single cell (Richer et al., 2013; Slifka and

Whitton, 2001) level. Individual CD8+ T cell clones become

more sensitive to their cognate antigen when they encounter in-

flammatory cytokines such as type I interferons and interleukin

(IL)-12 (Richer et al., 2013). Exposure to inflammatory cytokines

enhances the TCR signal transduction capacity of CD8+ T cells,

thereby reducing the threshold of antigen necessary for the in-

duction of effector functions such as the production of cytokines

and cytolysis (Richer et al., 2013). This represents an important

regulatory loop that couples the presence of inflammatory cyto-

kines to the activation of T cells, which likely limits immunopa-

thology. However, because the antigen sensitivity of CD8+

T cells is modulated by extrinsic factors, these regulatory circuits

could potentially favor persistence if they are inappropriately

activated during infection. It is currently unknown whether the

Immunity 48, 1–14, February 20, 2018 ª 2018 Elsevier Inc. 1

Page 3: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

A

0

25

50

75

100

125

-12 -11 -10 -9 -8

LCMV ArmLCMV cl13LCMV cl13

WT Host

Il10-/- Host

LCMV ArmLCMV cl13LCMV cl13LCMV cl13

shIl10ra

0

1

2

3

4

LCMVArm

LCMV

WTHost

Arm

0

25

50

75

100

125

-12 -11 -10 -9 -8

LCMV ArmLCMV Arm Il10-/- Host

WT Host

C

E

G

D

F

H

0 103 104 1050

20

40

60

80

100

% o

f Max

PD-1

LCMV cl13LCMV cl13

LCMV Arm

Il10-/- Host

WT Host

Isotype

% o

f Max

(IFN

-γ+ )

Log GP33(M)

B

0

1

2

LCMVArm

LCMVcl13

WTHost

Il10-/-

Host

3

4********

LCMVcl13

EC

50 (M

) (x1

0-10 )

% o

f Max

(IFN

-γ+ )

Log GP33(M)

EC

50 (M

) (x1

0-11 )

***

0

1

2

3

4

PD

-1 g

MFI

(x10

3 )

LCMVArm

LCMVcl13

WTHost

LCMVcl13

0

25

50

75

100

125

-12 -11 -10 -9 -8

% o

f Max

(IFN

-γ+ )

Log GP33(M)

0

1

2

3

shIl10ra

*** ****

*

EC

50 (M

) (x1

0-10 )

LCMVArm

LCMVcl13

LCMVcl13

LCMVcl13

AKT

pAKT

ERK1/2

pERK1/2

WT Host

LCMV Arm LCMV cl13

ZAP-70

pZAP-70

0 2 5CD3ε antibody

Time (min): 0 2 5 0 2 5

LCMV cl13

pPLCγ

PLCγ

ERK1/2

pERK1/2

WT Host

LCMV Arm LCMV cl13

0 2 5 0 2 5 0 2 5

LCMV cl13PMATime (min):

I J

Mock Transduced

shFFluc

MockTransduced

shFFluc

Il10-/-

Host

Il10-/-

Host

Il10-/-

HostIl10-/-

Host

(legend on next page)

2 Immunity 48, 1–14, February 20, 2018

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

Page 4: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

antigen sensitivity of CD8+ T cells is modulated in the context of

chronic viral infections or how this might be regulated.

Chronic viral infections induce an inflammatory milieu that

is distinct from those established by acute viral infections

(Ng and Oldstone, 2014b). These differences include changes

in both the duration of the inflammatory period and cytokines

produced. Several chronic viral infections, including HIV and

HCV, are associated with sustained induction of IL-10

in humans (Mannino et al., 2015). IL-10 is a broad spectrum

immunoregulatory cytokine that can inhibit the function of a

variety of immune cells and plays a critical role in dampening in-

flammatory responses (Corinti et al., 2001; Moore et al., 2001).

Increased serum detection of IL-10 coincides with disease

progression in patients with active HIV infection (Kwon and

Kaufmann, 2010). Additionally, elevated IL-10 signatures at

early time points following HCV challenge is associated with

chronic progression of the disease (Flynn et al., 2011). Thus,

the induction of IL-10 is a common feature of pathogens that

can establish chronic infections.

The role of IL-10 during the establishment of viral persistence

is clearly supported in mouse models of chronic viral infection.

Infection of mice with lymphocytic choriomeningitis virus strain

clone 13 (LCMV cl13) establishes chronic infection and induces

heightened and sustained production of IL-10, compared to

acute infection with the closely related LCMV Armstrong (Arm)

strain. The induction of IL-10 plays an important role in the estab-

lishment of persistence by LCMV cl13 as genetic deficiency of

IL-10- or antibody-mediated blockade of the IL-10 receptor

alpha chain (IL-10Ra) results in enhanced viral control (Brooks

et al., 2006; Ejrnaes et al., 2006). The mechanisms linking the

production of IL-10 to the establishment of chronic infection

and the cellular targets of IL-10 in this model remain major

knowledge gaps.

We have previously shown that pro-inflammatory cytokines

impact the antigen sensitivity of CD8+ T cells (Richer et al.,

2013). Thus, we tested the hypothesis that the anti-inflammatory

cytokine IL-10 induced during chronic infection increases the

threshold of antigen necessary for CD8+ T cell activation, thereby

enabling the pathogen to outpace the immune response and

establish persistence. We observed that IL-10 induced in

the context of LCMV cl13 triggered an N-glycan branching-

Figure 1. LCMV cl13 Infection Reduces the Antigen Sensitivity of CD8

Signaling

(A) Percentage of IFN-g+ P14 cells at day 8 after infection with LCMV Arm or LCM

titrated concentrations of GP33-41 peptide. Data (Mean ± SEM) are normalized to

(B) Summary (mean ± SEM) of EC50 for IFN-g+ P14 cells.

(C) Antigen sensitivity as in (A) at day 8 after infection with LCMV Arm in the indi

(D) Summary (mean ± SEM) of EC50 for IFN-g+ P14 cells.

(E) Representative histograms of expression of PD-1 on P14 cells at day 8 after

control.

(F) Summary (mean ± SEM) of geometric mean fluorescence intensity (gMFI) of P

(G) Antigen sensitivity as in (A) for P14 cells mock transduced or expressing shR

(H) Summary (mean ± SEM) of EC50 for IFN-g+ P14 cells.

(I) Immunoblot analysis of P14 cell lysates at day 8 after infection with indicat

crosslinking for the indicated time and equivalent amounts of total protein loade

(J) Same as (I), but using PMA stimulation.

Data in (A)–(H) represent three mice per group and are representative of at least tw

way ANOVA with Tukey’s post hoc analysis of multiple comparisons. *p < 0.05, **

least two independent experiments from three pooled mice per group. See also

mediated immunoregulatory loop that was associated with

decreased CD8+ T cell antigen sensitivity, thereby facilitating

the establishment of chronic infection.

RESULTS

IL-10 Directly Reduces the Antigen Sensitivity ofAntigen-Specific CD8+ T CellsWe previously showed that cytokines regulate the antigen sensi-

tivity of CD8+ T cells (Richer et al., 2013). As chronic viral infec-

tions establish a distinct inflammatory milieu that is character-

ized by the induction of IL-10 (Brooks et al., 2006; Ejrnaes

et al., 2006; Parish et al., 2014), we asked whether a virus that

establishes chronic infections negatively regulates the antigen

sensitivity of CD8+ T cells to counter immune defenses. To

address this question, we transferred congenically marked P14

TCR transgenic T cells into wild-type (WT) or IL-10-deficient

hosts (Il10�/�). CD8+ T cells from this transgenic mouse line ex-

press a TCR specific to GP33-41 (an immunodominant epitope of

LCMV), allowing us to measure the influence of the inflammatory

milieu on a population sharing identical TCRs and therefore iden-

tical affinity for their cognate antigen. Following adoptive trans-

fer, mice were infected with either LCMV Armstrong (Arm) or

LCMV clone 13 (cl13) to induce either acute or chronic infection,

respectively, and antigen sensitivity was measured at day 8

post-infection (Richer et al., 2013).

Infection with LCMV cl13 impaired the antigen sensitivity of

effector P14 cells compared to effector P14 cells from mice

infected with LCMV Arm (Figures 1A, 1B, S1A, and S1B).

Compared to P14 cells from LCMV cl13-infected mice, the anti-

gen sensitivity of P14 cells from LCMV Arm-infected mice was

higher based on the peptide concentration required to induce

50% of the maximum interferon (IFN)-g or tumor necrosis factor

(TNF)-a production (effective concentration 50 [EC50]) (Figures

1B and S1B). In the absence of IL-10, the antigen sensitivity of

P14 cells from LCMV cl13-infected mice was completely

restored (Figures 1A, 1B, S1A, and S1B). Conversely, IL-10 defi-

ciency had no effect on antigen sensitivity of effector P14 cells

during LCMV Arm infection (Figures 1C and 1D). In addition,

absence of IL-10 during LCMV cl13 infection resulted in an in-

crease in the number of P14 cells recovered as well as the

+ T Cells in an IL-10-Dependent Manner by Impairing Proximal TCR

V cl13 in the indicated host was determined following ex vivo stimulation with

the proportion of IFN-g+ cells at saturating peptide concentration (10 nM).

cated host.

infection with indicated strain of LCMV. Shaded histogram represents isotype

D-1 expression on P14 cells.

NAs targeting FFluc or Il10ra.

ed strains of LCMV from the indicated host. Cells were stimulated by CD3-

d into each lane. Total and phospho(p)-proteins were probed, as indicated.

o independent experiments. Data in (B), (D), (F), and (H) were analyzed by one-

p < 0.01, ***p < 0.001, ****p < 0.0001. Data in (I) and (J) are representative of at

Figures S1 and S2.

Immunity 48, 1–14, February 20, 2018 3

Page 5: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

percentage of these cells producing IFN-g and TNF-a (Figures

S1C–S1E). Further, we observed that P14 cells harvested from

LCMV cl13-infected mice had a reduced capacity to kill target

cells coated with low concentrations of peptide and this was

restored in the absence of IL-10 (Figure S1F). The antigen sensi-

tivity of P14 cells was also rescued via anti-IL-10R antibody

blockade during LCMV cl13 infection in WT mice (Figures S1G

and S1H). Thus, infection with LCMV cl13 reduced the antigen

sensitivity of CD8+ T cells in an IL-10-dependent manner.

During established chronic infections, persistent TCR stimula-

tion leads to T cell exhaustion, a hypo-functional state where

T cells fail to respond to antigen and express a high level of inhib-

itory receptors (Yi et al., 2010). At day 8 post-infection, P14 cells

from LCMV cl13-infected mice maintained heightened surface

expression of the co-inhibitory receptors PD-1, Lag-3, CTLA-4,

and Tim-3 irrespective of the presence of IL-10 (Figures 1E,

1F, and S1I–S1N). Thus, the antigen sensitivity of CD8+ T cells

can be restored in the absence of IL-10 independently of

changes in the expression of inhibitory receptors, further sug-

gesting that IL-10 and inhibitory receptors can induce T cell

dysfunction through distinct mechanisms (Brooks et al., 2008).

Although CD8+ T cells express the IL-10 receptor, the direct

effects of IL-10 on CD8+ T cells remain poorly defined. While

our experiments so far suggested a role for IL-10 in restricting

the antigen sensitivity of CD8+ T cells, they did not allow us to

determine whether this occurred through direct or indirectmech-

anisms. Thus, we asked whether inhibition of IL-10 signaling

specifically on CD8+ T cells was sufficient to restore antigen

sensitivity during LCMV cl13 infection. To investigate this, we

employed retroviral transduction to express short hairpin (sh)

RNAs targeting the Interleukin-10 receptor subunit alpha (Il10ra)

or the irrelevant gene firefly luciferase (shIl10ra and shFFluc,

respectively). Consistent with our previous results, knockdown

of the Il10ra gene (but not transduction with the irrelevant shRNA

targeting FFluc) restored CD8+ T cell antigen sensitivity during

LCMV cl13 infection (Figures 1G and 1H), supporting that IL-10

can regulate antigen sensitivity in a CD8+ T cell-intrinsic manner.

IL-10 Impairs the TCR Signal Transduction Capacity ofCD8+ T CellsWe have previously shown that inflammatory cytokines can

impact antigen sensitivity by increasing the efficiency of signal

transduction downstream of the TCR (Richer et al., 2013). There-

fore, we asked whether IL-10 antagonizes TCR signal transduc-

tion leading to the increased antigen threshold required for

activation. TCR ligation induces rapid phosphorylation of the

kinase Zap-70 and activation of PLCg, leading to the eventual

phosphorylation of AKT and ERK1/2, distal kinases activated

downstream of the signaling cascade (Smith-Garvin et al.,

2009). Tomeasure these signaling events at day 8 post-infection,

we stimulated enriched P14 cells by CD3ε-crosslinking to induce

activation in the absence of antigen presenting cells (APCs). P14

cells isolated from LCMV cl13-infected mice demonstrated a

defect in the activation of these signaling pathways despite no

changes in overall protein expression (Figure 1I). P14 cells

isolated from IL-10-deficient mice infected with LCMV cl13

exhibited restored TCR signaling capacity (Figure 1I). Further,

P14 cells from LCMV cl13-infected mice showed no defect in

ERK1/2 activation following stimulation with phorbol myristate

4 Immunity 48, 1–14, February 20, 2018

acetate (PMA; Figure 1J), which bypasses the proximal signaling

machinery. These data showed that P14 cells from LCMV cl13-

infected mice maintain the capacity for robust MAP kinase acti-

vation when the TCR proximal signaling machinery is bypassed.

This defect in signal transduction is not associated with a change

in the expression of surface components of the TCR or costimu-

latory molecules, as we observed that infection with LCMV cl13

did not alter the surface expression of the alpha (Va2) and beta

(Vb8.1) chains of the TCR, CD3ε, co-receptor CD8 or the costi-

mulatory molecule CD28 (Figure S2). Together, these data

demonstrated that infection with LCMV cl13 induces an IL-10-

dependent TCR-proximal signaling defect that is associated

with decreased antigen sensitivity.

IL-10 Impairs TCR:CD8 Co-localization during LCMVcl13 InfectionT cell activation requires the clustering of several membrane

proteins to induce effective and robust signal transduction. The

TCR and co-receptor CD8 exist in distinct lipid microdomains

within the T cell plasma membrane (Demotte et al., 2008).

Upon interacting with antigen, large-scale re-organization of

the plasma membrane allow for both TCR and CD8 to interact

with peptide:MHC class I on the APCs leading to signal trans-

duction. The interaction between TCR and the CD8 co-receptor

increases the affinity for peptide:MHC complex leading to

increased antigen sensitivity even when T cells are stimulated

in absence of APCs through CD3 cross-linking (Borger et al.,

2014; Cawthon and Alexander-Miller, 2002).

Because the capacity of the TCR to interact with the CD8 co-

receptor and to form micro-clusters is integral to dictating anti-

gen sensitivity, we hypothesized that reduced TCR and CD8

co-receptor association is linked to a decrease in CD8+ T cell an-

tigen sensitivity. To test whether TCR:CD8 co-localization is

impaired during LCMV cl13 infection, we used a well-defined

flow cytometry adapted Fӧrster resonance energy transfer

(FRET) approach (Figure 2A; Demotte et al., 2008). On day 8

post-infection with LCMV Arm, we observed robust co-localiza-

tion of TCR and CD8 on P14 cells and this was reduced on P14

cells isolated from LCMV clone 13-infectedmice (Figures 2B and

S3A). TCR and CD8 co-localization was rescued in P14 cells iso-

lated from LCMV cl13-infected Il10�/� mice (Figures 2B and

S3A), correlating with their restored TCR signaling capacity

and antigen sensitivity (Figure 1). These data suggested that

infection with LCMV cl13 can lead to IL-10-induced impairment

of the capacity of CD8+ T cells to co-localize the TCR and CD8

co-receptors and this is correlated with reduced T cell respon-

siveness to antigen.

LCMV cl13 Infection Increases N-glycan Branching onT Cells in an IL-10-Dependent MannerThe expression of Mgat5, a Golgi-resident glycosyltransferase,

has been shown to restrict TCR clustering by increasing the

branching of N-glycans on the surface of T cells at steady state

(Demetriou et al., 2001). Mgat5-modified N-glycans can be

further extended resulting in high-affinity binding sites for the ga-

lectin family of soluble b-galactoside binding lectins (Figure 2C;

Ilarregui et al., 2005). Galectin binding of surface glycoproteins

forms a lattice that restricts protein diffusion due to multivalent

interactions with multiple glycoproteins (Elola et al., 2015;

Page 6: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

A

0

20

40

60

80

FRET

Uni

t s

********

B

Galectinbinding

Mgat5

Man

GlcNAc

Gal

Sialic acid

PHA-Lbinding

N-X-S/T

C

F

0

1

2

3

4

5 **

0

1

2

3

4**

K

PHA-L

E%

of M

a x

0 102 103 104 1050

20

40

60

80

100

J

Gal3

% o

f Max

0 10 10 10 102 3 4 50

20

40

60

80

100

Naïve P. yoelii- GP33

01234567

Mga

t5 R

elat

ive e

xpre

ssio

n ****H

D

P14Naïve

0

5

10

15 ******** ****

Mga

t5 R

elat

ive

expr

essi

on

G I

MG

AT5

Rel

ativ

ee x

pre s

sion

HCVNaïve

HCV chronic

0

2

4

6

8

10

12 *

LCMV cl13LCMV cl13

LCMV Arm

Il10-/- Host

WT Host

10 nm

TCR CD8

FMO

LCMVArm

LCMVcl13

WTHost

LCMVcl13

LCMVArm

LCMVcl13

WTHost

LCMVcl13

PH

A-L

gM

FI (x

102 )

LCMVArm

LCMVcl13

WTHost

LCMVcl13

LCMVArm

LCMVcl13

WTHost

PHA-L pull down

Whole cell lysate

TCR α/β

TCR α/β

LCMVcl13

Gal

3 gM

FI (x

102 )

LCMVArm

LCMVcl13

WTHost

LCMVcl13

Il10-/-

Host

Il10-/-

Host

Il10-/-

Host

Il10-/-

Host

Il10-/-

Host

Figure 2. IL-10 Restricts the Co-localization of TCR with CD8 Co-receptor and Enhances N-glycan Branching of CD8+ T Cells

(A) Schematic of FRET approach.

(B) Summary (mean ± SEM) of FRET units for P14 cells at day 8 after infection with indicated strain of LCMV in the indicated host.

(C) Schematic of Mgat5-modified N-glycan and respective galectin and PHA-L binding sites.

(D) Relative Mgat5 expression in day 8 effector P14 cells from indicated conditions compared to naive P14 cells (mean ± SEM).

(E) Representative histogram of PHA-L binding on effector P14 cells at day 8 after infection. Shaded histogram represents fluorescenceminus one (FMO) control.

(F) Summary (mean ± SEM) of gMFI of PHA-L binding.

(G) Immunoblot of a/b TCR after pull-down with PHA-L-conjugated beads.

(H) Relative Mgat5 expression in day 7 P14 cells after infection with P. yoellii-GP33 compared to naive P14 cells (mean ± SEM).

(I) Relative MGAT5 expression in total CD8+ T cells isolated from human patients with chronic HCV infection compared to a naive cohort (mean ± SEM).

(J) Representative histogram of Gal3 binding on P14 cells at day 8 after infection with indicated strain of LCMV. Shaded histogram represents FMO control. Refer

to (E) for color legend.

(K) Summary (mean ± SEM) of gMFI of Gal3 binding on P14 cells.

Data in (B), (D)–(F), and (H)–(K) represent threemice per group and are representative of at least two experiments. Data in (G) are from at least two pooledmice per

group and are representative of three experiments. Data in (I) are from five patients per group. Data in (B), (D), (F), and (K) were analyzed by one-way ANOVA with

Tukey’s post hoc analysis of multiple comparisons. Data in (H) and (I) were analyzed by two-tailed unpaired t test. *p < 0.05, ****p < 0.0001. See also Figure S3.

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

Ilarregui et al., 2005). However, whether this is amechanism gov-

erning CD8+ T cell activation and whether this can be modulated

by IL-10 remains unknown. Thus, we asked whether Mgat5

expression is linked to IL-10-mediated regulation of CD8+

T cell antigen sensitivity. At day 8 after infection, we observed

an 11-fold increase in Mgat5 transcript expression in CD8+

T cells responding LCMV cl13 infection relative to naive T cells

(Figure 2D) while the expression of other glycosyltransferases

Mgat1, Mgat2, and Mgat4 remained unchanged (data not

shown). Similarly, we observed an IL-10-dependent increase

in Il10ra transcript expression by P14 cells during LCMV cl13

infection (Figure S3B). Increased Mgat5 expression was not

observed in CD8+ T cells responding to LCMV Arm infection or

by CD8+ T cells responding to LCMV cl13 infection in Il10�/�

mice, establishing that IL-10 expression during LCMV cl13

infection regulates Mgat5 expression (Figure 2D). To deter-

mine whether increased Mgat5 expression is associated with

enhanced N-glycan branching on CD8+ T cells, P14 cells were

stained with PHA-L, a lectin isolated from Phaseolus vulgaris

that specifically binds to Mgat5-modified branched N-glycans

(Figure 2C; Demetriou et al., 2001). We observed increased bind-

ing of PHA-L on CD8+ T cells frommice infected with LCMV cl13

Immunity 48, 1–14, February 20, 2018 5

Page 7: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

infection and this increase was dependent on IL-10 (Figures 2E

and 2F). Mgat5-mediated glycosylation facilitates binding of

galectin 3 (Gal3) to the TCR, thereby limiting its redistribution

(Demetriou et al., 2001). Thus, we asked whether Mgat5-modi-

fied glycans were detectable on the TCR specifically. At day 8 af-

ter infection, we precipitatedMgat5-modified glycoproteins from

whole P14 cell lysates using PHA-L-conjugated agarose beads.

PHA-L beads precipitated detectable TCR only from T cells from

WT mice infected with LCMV cl13 and not from LCMV Arm-in-

fected WT mice or LCMV cl13-infected Il10�/� mice (Figure 2G).

Thus, Mgat5-mediated glycosylation of the TCR was enhanced

during LCMV cl13 infection in an IL-10-dependent manner.

Chronic Plasmodium and HCV Infections RegulateExpression of Mgat5

To address whether other pathogens that establish chronic

infection can also regulate the expression of Mgat5 in CD8+

T cells during the early stages of infection, we used a murine

model of chronic parasitic infection. We adoptively transferred

congenically marked P14 cells and subsequently infected mice

with Plasmodium yoelii (a parasite that induces sustained IL-10

production and stimulates T cell exhaustion) (Butler et al.,

2011; Kobayashi et al., 1996) expressing the model antigen

GP33-41 of LCMV. At day 7 after infection, we observed increased

expression of Mgat5 and Il10ra compared to naive P14 cells

(Figures 2H and S3C). Thus, these data suggested that the in-

duction of Mgat5 may represent a conserved mechanism that

limits T cell responses and favors the establishment of persistent

or prolonged infections.

We next asked whether this mechanism of regulation is also

observed during chronic viral infections in humans. To address

this, we examined the expression of MGAT5 and IL10RA in

CD8+ T cells isolated from peripheral blood of subject with

chronic infection HCV (a chronic viral infection that has also

been associated with the induction of IL-10 [Frebel et al.,

2010]). We observed that CD8+ T cells from subjects chronically

infected with HCV expressed higher levels of MGAT5 and

IL10RA compared to CD8+ T cells from HCV naive individuals

from the same cohort (Figures 2I and S3D). Therefore, our data

showed that chronic infections in humans also result in increased

MGAT5 expression by CD8+ T cells.

Galectin Inhibition Restores CD8+ T Cell AntigenSensitivityWhile we observed an increase in Mgat5 expression and

N-glycan branching, this would not be sufficient to restrict sur-

face receptor interaction unless the N-glycans are engaged to

form a restrictive network. The galectin protein family has several

diverse members that are ubiquitously expressed in both the

intracellular and extracellular environment. Previous studies

demonstrate that in T cells, Gal3 is the predominant galectin

involved in restricting TCR clustering (Demetriou et al., 2001).

To investigate whether Gal3 binding could explain the impaired

TCR:CD8 co-receptor association during LCMV cl13 infection,

we measured extracellular Gal3 binding on CD8+ T cells

by flow cytometry. Consistent with our previous results, we

observed increased Gal3 binding on the surface of CD8+

T cells during LCMV cl13 infection that corresponds with

enhanced PHA-L binding and is lost in the absence of IL-10 (Fig-

6 Immunity 48, 1–14, February 20, 2018

ures 2E, 2F, 2J, and 2K). As staining was performed directly

ex vivo (without addition of exogenous Gal3), these data showed

that CD8+ T cells bindmore Gal3 in vivo during the establishment

of chronic infection and that this can be regulated by IL-10.

Furthermore, Gal3 expression was not increased at the tran-

script level in either P14 cells or total splenocytes or at the

protein level in the serum of either LCMV Arm- or LCMV cl13-in-

fected mice (Figures S3E–S3G). Together these data suggested

that increased N-glycan branching, rather than changes in Gal3

expression, underlies the increased binding of Gal3 to the sur-

face of CD8+ T cells.

To identify a functional role of galectin binding in reducing

CD8+ T cell antigen sensitivity and TCR signaling, we asked

whether broad inhibition of galectin binding using the competi-

tive inhibitor D-Lactose is sufficient to restore CD8+ T cell func-

tion. Ex vivo treatment with D-Lactose was sufficient to remove

bound Gal3 from the surface of P14 cells (Figures S4A and

S4B) without reducing the surface expression of the co-inhibitory

receptor PD-1 (Figures S3C and S3D). When comparing the

function of P14 cells isolated from the same LCMV cl13-infected

donor mice, we observed that the addition of D-Lactose during

stimulation with peptide restored CD8+ T cell antigen sensitivity

(Figures 3A and 3B). Similarly, galectin inhibition restored

TCR:CD8 membrane co-localization (Figures 3C and S4E) and

rescued the TCR signaling capacity of CD8+ T cells (Figure 3D).

These data demonstrated that inhibiting galectin binding can

rescue IL-10-mediated CD8+ T cell suppression. Treatment

with D-Lactose had no effect on P14 cells isolated from LCMV

Arm-infected mice (Figures 3E and 3F). Together, these data

supported a model in which the inhibitory effects of IL-10 on an-

tigen sensitivity and signal transduction are mediated by the

binding of galectins on the surface of T cells.

Gal3 Deficiency Restores Antigen Sensitivity and ViralControl Despite Increased N-glycan BranchingOur data showed that broad galectin inhibition with the compet-

itive inhibitor D-Lactose rescued antigen sensitivity and TCR

signaling capacity (Figure 3). Because Gal3 is believed to be

the dominant lectin involved in restricting TCR diffusion, we

asked whether Gal3 plays a role in IL-10-mediated CD8+ T cell

dysfunction during LCMV cl13 infection. To address this, we

adoptively transferred congenically marked WT P14 cells

into Gal3-deficient mice (Lgals3�/�) and subsequently infected

them with LCMV cl13. At day 8 after infection, we observed

that the absence of Gal3 restored the antigen sensitivity of P14

cells with an EC50 comparable to that of P14 cells from LCMV

Arm-infected WT mice (Figures 4A and 4B). The absence of

Gal3 did not impact PD-1 expression, which remained high

during LCMV cl13 infection, further suggesting that the effects

of IL-10 on T cell function are independent of PD-1 expression

(Figures S5A and S5B). In addition, reduced Gal3 binding on

the surface of CD8+ T cells restored TCR:CD8 co-localization

(Figures 4C and S5C–S5E) despite elevated PHA-L binding

that was equivalent on P14 cells recovered from both WT and

Lgals3�/� mice infected with LCMV cl13 (Figures 4D and 4E).

This suggested that, even in the absence of Gal3, LCMV cl13

infection induced an increase in Mgat5-modified N-glycan

branching (as indicated by elevated PHA-L binding), but that

binding of host-derived Gal3 was necessary to restrict T cell

Page 8: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

BA

LCMVArm

LCMVcl13

LCMVcl13

+ Lactose

FRET

Uni

ts

0

20

40

60

80

100 ********

LCMV ArmLCMV cl13LCMV cl13 + Lactose

C

D

-11 -10 -9 -80

25

50

75

100

125

-12

% o

f Max

(IFN

-γ+ )

Log GP33(M)

0

1

2

3

4

********

EC

50 (M

) (x1

0-10 )

LCMVArm

LCMVcl13

LCMVcl13

+ Lactose

+ Lactose

LCMV Arm LCMV cl13

0 2 5CD3ε antibody

Time (min): 0 2 5 0 2 5

LCMV cl13

AKT

pAKT

ERK1/2

pERK1/2

ZAP-70

pZAP-70

pPLCγ

PLCγ

LCMV ArmLCMV Arm + Lactose

LCMVArm

LCMVArm

+ Lactose

0

2

4

6

8

10

-12 -11 -10 -9 -80

25

50

75

100

125

E F

% o

f Max

(IFN

-γ+ )

Log GP33(M)

EC

50 (M

) (x1

0-11 )

Figure 3. Inhibition of Galectin Binding Rescues T Cell Function during LCMV cl13 Infection

(A) Antigen sensitivity as in Figure 1A of P14 cells from day 8 after infection with the indicated strain of LCMV in WT hosts. Cells were stimulated with or without

50 mM D-Lactose.

(B) Summary (mean ± SEM) of EC50 for IFN-g+ P14 cells.

(C) Summary FRET units (mean ± SEM) with or without 50 mM D-Lactose treatment.

(D) Immunoblot analysis as in Figure 1I following treatment with or without 50 mM D-Lactose.

(E) Antigen sensitivity in P14 cells isolated from LCMV Arm infection with or without treatment with D-Lactose as in Figure 1A.

(F) Summary (mean ± SEM) EC50 for IFN-g+ P14 cells.

Data in (A)–(C), (E), and (F) represent three mice per group and are representative of at least three experiments. Data in (D) represent total protein of three pooled

mice per group and is representative of at least two experiments. Data in (B) and (C) were analyzed by one-way ANOVAwith Tukey’s post hoc analysis of multiple

comparisons. Data in (F) were analyzed by two-tailed unpaired t test. ****p < 0.0001. See also Figure S4.

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

function. Together, these data suggested that Gal3 binding to

glycoproteins on the surface of T cells plays a central role in

mediating the IL-10-induced reduction in CD8+ T cell antigen

sensitivity.

The Lgals3�/� mouse model allowed us to determine

whether restoring CD8+ T cell antigen sensitivity leads to an

improvement in the capacity of the host to control viral infec-

tion. Genetic deletion of Gal3 dramatically improved viral

control with some mice clearing LCMV cl13 infection by

day 8 and others exhibiting approximately 2 log reductions in

viral titers in both the kidneys and the liver (Figures 4F and

4G). Enhanced virus control in Lgals3�/� mice was not simply

due to an increase in CD8+ T cell numbers, as T cell expansion

was reduced relative to P14 cells responding to LCMV Arm

infection (Figure S5F). Thus, despite increased N-glycan

branching on T cells responding to LCMV cl13 in the presence

of IL-10, Gal3 deficiency was associated with restored CD8+

T cell antigen sensitivity and increased capacity of the host to

control viral infection.

Our previous experiments suggested a central role for IL-10 in

increasing N-glycan on CD8+ T cells, thereby facilitating Gal3-

mediated repression of T cell function and host protection. To

investigate whether Mgat5 mediates these effects, we trans-

duced P14 cells with retroviral constructs expressing shRNAs

targetingMgat5 or the irrelevant gene Firefly luciferase (shMgat5

and shFFluc, respectively). At day 8 post-infection with LCMV

cl13, Mgat5 knockdown in P14 cells restored antigen sensitivity

independently of any changes in the surface expression of

PD-1 (Figures 5A–5D). Mgat5 knockdown also decreased Gal3

binding and PHA-L staining on CD8+ T cells, showing that the

reduction of Mgat5 expression decreased N-glycan branching

(Figures 5E–5H). Conversely, transduced P14 cells expressing

the control shRNA showed similar EC50s and similar binding

of PHA-L and Gal3 compared to mock transduced cells (Fig-

ure 5). Collectively, these data supported that the IL-10-medi-

ated increase in Mgat5 expression is important to modify the

glycosylation pattern on CD8+ T cells and is associated with

T cell dysfunction during LCMV cl13 infection.

IL-10 Regulates Mgat5 Expression and EnhancesN-glycan BranchingTo address whether IL-10 directly regulates the expression

of Mgat5 and alters glycosylation and investigate why this regu-

latory loop is engaged only in the context of chronic infection, we

established an ex vivo system. Infection with LCMV cl13 induces

increased and sustained IL-10 expression compared to LCMV

Immunity 48, 1–14, February 20, 2018 7

Page 9: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

A BFR

ET U

nits

0

20

40

60

80********

C

PHA

-LgM

FI (x

10)

0

1

2

3

4 ****E

-12 -11 -10 -9 -80

25

50

75

100

125

D

F

0

0.5

1.0

1.5

2.0

2.5 ********

LCMV ArmLCMV cl13LCMV cl13

WT Host

Lgals3-/- Host

LCMV cl13LCMV cl13

LCMV Arm

Lgals3-/- Host

WT Host

FMO

% o

f Max

(IFN

-γ+ )

Log GP33(M)

EC

50 (M

) (x1

0-10 )

LCMVArm

LCMVcl13

WTHost

LCMVcl13

LCMVArm

LCMVcl13

WTHost

LCMVcl13 PHA-L

0 102 103 104 1050

20

40

60

80

100

% o

f Max

LCMVArm

LCMVcl13

WTHost

LCMVcl13

6n.d.

5n.d.

L.O.D. L.O.D.

LCMVArm

LCMVcl13

WTHost

LCMVcl13

6n.d.

2n.d.

LCMVArm

LCMVcl13

WTHost

LCMVcl13

G

PFU

/gki

dney

(Log

10)

2

3

4

5

6

7

8

PFU

/gliv

er( L

og10

)

2

3

4

5

6

7

8

Lgals3-/-

Host

Lgals3-/-

Host Lgals3-/-

Host

Lgals3-/-

Host Lgals3-/-

Host

Figure 4. Gal3 Regulates Antigen Sensitivity and Limits T Cell Function during LCMV cl13 Infection

(A) Antigen sensitivity as in Figure 1A for P14 cells at day 8 after infection with the indicated LCMV strain in the indicated host.

(B) Summary (mean ± SEM) EC50 for IFN-g+ P14 cells.

(C) Summary (mean ± SEM) FRET units.

(D) Representative histograms of PHA-L binding on P14 cells. Shaded histogram represents FMO control.

(E) Summary (mean ± SEM) gMFI of PHA-L binding.

(F and G) Plaque forming units (PFUs) per gram of indicated organ at day 8 after infection with indicated strains of LCMV in the indicated host.

Data in (A)–(E) represent three mice per group and are representative of two experiments. Data in (F) and (G) represent six mice per group pooled from two

experiments. Data in (B), (C), and (E) were analyzed by one-way ANOVA with Tukey’s post hoc analysis of multiple comparisons. **p < 0.01, ****p < 0.0001. See

also Figure S5.

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

Arm infection (Brooks et al., 2006; Frebel et al., 2010). Thus, we

tested whether dose and timing of exposure to IL-10 are impor-

tant for the regulating ofMgat5 expression and N-glycan branch-

ing. We observed that expression of Mgat5 and increased

N-glycan branching were dependent on the dose of IL-10 (Fig-

ures 6A–6C). Similarly, while short-term treatment with IL-10

(day 1 or day 1 and 2 of culture only) resulted in modestly

increased expression of Mgat5 and N-glycan branching, sus-

tained exposure to IL-10 (all days of culture) resulted in much

greater expression of both (Figures 6D–6F). Thus, both the

dose and the duration of exposure to IL-10 were critical for the

8 Immunity 48, 1–14, February 20, 2018

regulation of N-glycan branching, which likely explains why this

immunoregulatory loop is strongly engaged during LCMV cl13

but not LCMV Arm infection.

To investigate whether IL-10 acts on CD8+ T cells through ca-

nonical STAT3-dependent signaling pathways, we employed a

previously described STAT3 inhibitor (WP1066) (Iwamaru et al.,

2007). We observed upregulation ofMgat5 expression following

activation in the presence of IL-10, which was impaired by

STAT3 inhibition (Figure 6G). Furthermore, upregulation of

Mgat5 was associated with increased binding of PHA-L that

was lost upon inhibition of STAT3 (Figures 6H and S6A). Because

Page 10: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

B

LCMV ArmLCMV cl13LCMV cl13

FMO

LCMV cl13

0 103 104 1050

20

40

60

80

100

Gal3

% o

f Max

0 103 104 1050

20

40

60

80

100

% o

f Max

A

-12 -11 -10 -9 -80

25

50

75

100

125

C D

E F

0 102 103 104 1050

20

40

60

80

100

% o

f Max

0.0

0.5

1.0

1.5

2.0 *** *** **

0.0

0.5

1.0

1.5**** **** ****

*****G H

% o

f Max

(IFN

-γ+ )

Log GP33(M)

LCMV ArmLCMV cl13LCMV cl13

Mock TransducedshMgat5

LCMV cl13 shFFluc 0

1

2

3

4 *** *** ****

EC

50 (M

) (x1

0-10 )

LCMVArm

LCMVcl13

MockTransduced

shMgat5

LCMVcl13

LCMVcl13

shFFluc

PD-1

Mock TransducedshMgat5shFFluc 0

1

2

3

4

5*

***

LCMVArm

LCMVcl13

MockTransduced

shMgat5

LCMVcl13

LCMVcl13

shFFluc

PD

-1 g

MFI

(x10

2 )

Gal

3 gM

FI (x

103 )

LCMVArm

LCMVcl13

MockTransduced

shMgat5

LCMVcl13

LCMVcl13

shFFluc

PHA-L

PH

A-L

gM

FI (x

103 )

LCMVArm

LCMVcl13

MockTransduced

shMgat5

LCMVcl13

LCMVcl13

shFFluc

Figure 5. Mgat5 Knockdown Rescues CD8+ T Cell Antigen Sensitivity during LCMV cl13 Infection

(A) Antigen sensitivity as in Figure 1G for P14 cells expressing an shRNA targeting Mgat5, FFluc, or mock transduced.

(B) Summary (mean ± SEM) of EC50 for IFN-g+ P14 cells.

(C) Representative histograms of PD-1 expression, shaded histogram represents isotype control.

(D) Summary (mean ± SEM) gMFI of PD-1 expression on P14 cells.

(E) Representative histograms of Gal3 binding on P14 cells, shaded histogram represents FMO control. Refer to (C) for color legend.

(F) Summary (mean ± SEM) gMFI of Gal3 binding on P14 cells.

(G) Representative histograms of PHA-L binding on P14 cells, shaded histogram represents FMO control. Refer to (C) for color legend.

(H) Summary (mean ± SEM) gMFI of PHA-L binding on P14 cells. Data represent three mice per group and are representative of two experiments.

Data in (B), (D), (F), and (H) were analyzed by one-way ANOVA with Tukey’s post hoc analysis of multiple comparisons. *p < 0.05, **p < 0.01, ***p < 0.001,

****p < 0.0001.

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

Gal3 was ubiquitously expressed and its expression was

unchanged during LCMV infection (Figures S3E and S3F), sub-

strate (branched N-glycans) availability likely acts as the rate-

limiting step to galectin binding. Thus, we asked whether recom-

binant Gal3 supplied exogenously could recapitulate our in vivo

observations. Indeed, exogenous recombinant Gal3 only bound

to cells treated with IL-10 in the absence of STAT3 inhibitor (Fig-

ures 6I and S6B), supporting our model that Mgat5-mediated

glycan modifications is induced by IL-10 and acts as the limiting

step to galectin-mediated repression. Treatment with other cyto-

kines that signal through STAT3 such as IL-6 and IL-21 was not

sufficient to induce Mgat5 expression (Figure S6C). Together,

these data supported that IL-10 acted directly on CD8+ T cells

in a STAT3-dependent manner to induce Mgat5 expression,

which sensitized T cells to Gal3 binding.

Our ex vivo experiments suggested a regulatory pathway

through which IL-10 can inhibit CD8+ T cells by inducing

Mgat5 expression. Knockdown of Il10ra in P14 cells also resulted

in reduced binding of Gal3 and reduced Mgat5-modified

N-glycans (as measured by PHA-L) compared to mock-trans-

duced cells (Figures 6J–6K and S6D and S6E) and control

shRNA transduced cells following LCMV cl13 infection. Thus,

the composite of our data suggested a model in which IL-10

acted directly on CD8+ T cells to regulate N-glycan branching,

Immunity 48, 1–14, February 20, 2018 9

Page 11: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

H

Mga

t5R

elat

ive

expr

essi

on

G

0

10

20

30

40

***********

STAT3iIL-10 -

--+

+- +

+

0

2

4

6

8

************

PH

A-L

gM

FI (x

102 )

STAT3iIL-10 -

--+

+- +

+

0 3.125 6.25 12.5 25 50 100 2000

10

20

30

IL-10 (ng/mL)

% o

f Max

0 102 103 104 1050

20

40

60

80

100

101

PHA-L0 3.125 6.25 12.5 25 50 100 200

0

2

4

6

IL-10 (ng/mL)

PH

A-L

gM

FI (x

102 )

d1 d1+d2 All none0

1

2

3

4

5

PH

A-L

gM

FI (x

102 )

% o

f Max

0 102 103 104 1050

20

40

60

80

100

101

PHA-Ld1 d1+d2 All none

0

10

20

30****

**** ****

Mga

t5R

elat

ive

expr

essi

on

I

0

1

2

3

4*******

****

Gal

3 gM

FI (x

102 )

STAT3iIL-10 -

--+

+- +

+

0

2

4

6

8*** ***

***

Gal

3 gM

FI (x

102 )

LCMVArm

LCMVcl13

MockTransduced

shIl10ra

LCMVcl13

LCMVcl13

shFFluc

K

LCMVArm

LCMVcl13

MockTransduced

shIl10ra

LCMVcl13

LCMVcl13

shFFluc

0.0

0.5

1.0

1.5

**** ********

****

*

PH

A-L

gM

FI (x

102 )

d1d1+d2Allnone

FMO

100 50 25 12.56.253.125 0

200

IL-10 (ng/mL):

Mga

t5R

elat

ive

expr

essi

onB CA

D E F

J

FMO

****

********

********

****

****

*

****** *****

* **

Figure 6. IL-10 Directly Regulates Mgat5 Expression and Reduces Antigen Sensitivity in a T Cell-Intrinsic Manner

(A) Relative Mgat5 expression in CD8+ T cells activated ex vivo in the presence of increasing doses of IL-10 for 8 days compared to untreated (0) group

(mean ± SEM).

(B) Representative histograms of PHA-L binding to CD8+ T cells, shaded histogram represents FMO control.

(C) Summary (mean ± SEM) gMFI of PHA-L on CD8+ T cells following IL-10 treatment.

(D) RelativeMgat5 expression in CD8+ T cells activated ex vivo and cultured for 8 days with exposure to 200 ng/mL of IL-10 on the first day (d1), first and second

day (d1+d2), or all days of culture (All) compared to untreated (none) group (mean ± SEM).

(E) Representative histograms of PHA-L binding to CD8+ T cells for cells treated as in (D); shaded histogram represents FMO control.

(F) Summary (mean ± SEM) gMFI of PHA-L on CD8+ T cells following IL-10 treatment as in (D).

(G) RelativeMgat5 expression in CD8+ T cells activated ex vivo in the presence or absence of IL-10 and/or STAT3 inhibitor (WP1066) compared to untreated group

(mean ± SEM).

(H) Summary (mean ± SEM) gMFI of PHA-L on CD8+ T cells.

(I) Summary (mean ± SEM) gMFI of Gal3 binding on CD8+ T cells following incubation with exogenous Gal3.

(J) Summary (mean ± SEM) gMFI of Gal3 binding on P14 cells transduced or mock-transduced as indicated.

(K) Summary (mean ± SEM) gMFI of PHA-L binding on P14 cells transduced or mock-transduced as indicated. Data represent three mice per group and are

representative of two experiments.

Data in (A), (C), (D), and (F)–(K) were analyzed by one-way ANOVA with Tukey’s post hoc analysis of multiple comparisons. Data in (A) and (C) are indicated as

significant relative to untreated (0 ng/mL) group only. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. See also Figure S6.

10 Immunity 48, 1–14, February 20, 2018

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

Page 12: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

leading to increased Gal3 binding and reduced antigen sensi-

tivity contributing to T cell dysfunction during the establishment

of chronic viral infection.

DISCUSSION

While the impact of chronic infections on host immune function,

particularly the progressive development of T cell exhaustion,

has been well described (Wherry and Kurachi, 2015), the early

events allowing pathogens to establish persistence remain less

defined. Herein, we demonstrated that pathogens that establish

chronic infections triggered reductions in CD8+ T cell antigen

sensitivity and that this required CD8+ T cell-intrinsic IL-10

signaling. Mechanistically, our data supported a model in which

IL-10 signaling, via STAT3, increased the expression of the

glycosyltransferase Mgat5 leading to enhanced branched

N-glycans on glycoproteins (including the TCR) on the surface

of CD8+ T cells. Mgat5-modified N-glycans, in turn, served as

ligands for the binding of Gal3 and the formation of a restrictive

lattice that limited the capacity of TCR and the CD8 co-receptor

to interact, which was associated with a defect in TCR signal

transduction. This reduction in CD8+ T cell antigen sensitivity

increased the antigenic threshold required for T cell activation,

which may allow the rapidly replicating pathogen to outpace

the immune system and establish persistence. Together, our

data suggested a mechanism of CD8+ T cell regulation by anti-

inflammatory cytokines that act through post-translational modi-

fication networks to modulate TCR sensitivity and CD8+ T cell

function. In addition, our data revealed that this regulatory loop

is inappropriately activated during certain infections and is asso-

ciated with the establishment of persistence.

IL-10 is an important immunomodulatory cytokine that im-

pacts a variety of cell types. While the inhibitory role of IL-10

on APCs and CD4+ T cells is becoming well defined, much less

was known about its impact on CD8+ T cells (Ip et al., 2017;

Krawczyk et al., 2010). In fact, previous research shows that

IL-10 can have a stimulatory role for CD8+ T cells, aiding in tumor

regression and acting as a substitute for signal 3 in the absence

of IL-2 (Emmerich et al., 2012; Santin et al., 2000). Conversely,

our data supported that IL-10 can act directly on CD8+ T cell

to exert specific inhibitory effects. Similar paradoxical effects

of cytokines, such as type I interferon, have been documented

during chronic viral infections (Ng and Oldstone, 2014a; Wilson

and Brooks, 2011). Thus, the specific timing and inflammatory

context likely play an important role in the transcriptional network

and effects induced by IL-10 on CD8+ T cells. In fact, our data

supported that both the dose and duration of exposure to

IL-10 are critical to engage this regulatory loop, likely explaining

why it is engaged only during the establishment of chronic

infection.

N-glycosylation of the TCR is an established regulator of T cell

antigen sensitivity, as mutational approaches reducing N-glyco-

sylation can improve the sensitivity of T cells for their antigen

(Kuball et al., 2009). Mgat5 plays a central role in regulating the

activation of T cells by modulating the branching of N-glycans.

Previous studies demonstrate that Mgat5-deficient naive

T cells spontaneously cluster their TCR and have a lower anti-

genic threshold for activation at steady-state (Demetriou et al.,

2001). In addition, Mgat5-deficient mice develop spontaneous

autoimmunity and are more susceptible to the induction of

experimental encephalomyelitis (EAE), clearly highlighting the

importance of this enzyme as a gatekeeper of T cell activation

(Demetriou et al., 2001; Grigorian and Demetriou, 2011). Here,

we demonstrated that IL-10 signaling directly to CD8+ T cells

enhanced the expression of Mgat5 to negatively regulate CD8+

T cell function. This novel regulatory loop appears conserved

across several infections and in humans, as we observed that

CD8+ T cells from mice infected with Plasmodium yoelii (a

parasite that establishes chronic infection) and from patients

chronically infected with HCV express elevated levels of

Mgat5 transcripts. Our data established modulation of N-glycan

branching as a critical rate-limiting step of the Gal3-mediated

suppression of T cell function. This is supported by the observa-

tion that Gal3 expression is not regulated during LCMV infection

and by the inability of CD8+ T cells to bind exogenously provided

Gal3 unless they were stimulated in the presence of IL-10. One

remaining question is how IL-10 regulates the expression of

Mgat5. While our data supported that the increased expression

of Mgat5 depends on the canonical STAT3 signaling pathway,

there are no STAT3 consensus binding sites within the Mgat5

promoter region. One possibility is that STAT3 may complex

with ETS-1 to potentiate Mgat5 transcription as STAT3-ETS-1-

SP1 transcriptional complexes have been described, and

ETS-1 is a documented regulator of Mgat5 (Bian et al., 2011;

Kang et al., 1996). Alternatively, STAT3 signaling may lead to

the induction of other transcription factors leading to enhanced

Mgat5 expression. In support of this, STAT3 is known to direct

the expression and epigenetic modifications of a number of

transcription factors in T cells (Durant et al., 2010). Determining

how IL-10 regulates Mgat5 expression will be the subject of

future investigations.

The regulation of Mgat5 by IL-10 over the course of infection

may represent an immunoregulatory loop that plays an important

role even during a protective immune response. This may serve

to prevent some CD8+ T cells from becoming terminally differen-

tiated, thereby maintaining their capacity to develop into mem-

ory CD8+ T cells. In support of this, IL-10 plays an important

role in the development of memory CD8+ T cells following clear-

ance of infection (Cui et al., 2011). While some of the role of IL-10

in that context is to insulate the T cells from the effects of other

inflammatory cytokines, it is tempting to speculate that it may

also have a direct effect on CD8+ T cells by regulating Mgat5

expression and N-glycan branching. In the context of chronic in-

fections, the sustained and heightened production of IL-10 may

simply amplify this regulatory loop, leading to the inability of

T cells to respond efficiently, thereby favoring the establishment

of pathogen persistence.

Our data suggested that the galectin N-glycan lattice

dampens CD8+ T cell antigen sensitivity during chronic viral in-

fections and that this step is controlled by IL-10 signaling to

CD8+ T cells. We showed that disruption of this regulatory

pathway restored some T cell function during infection and is

associated with better viral control. Of note, tumor-derived

IL-10 plays an important role in immune evasion in some cancers

and local administration of galectin inhibitors can restore im-

mune detection and reduce tumor burden in these models (De-

motte et al., 2010; Sun et al., 2015). Therefore, pharmacological

inhibition of IL-10 orMgat5 regulatory circuits may also provide a

Immunity 48, 1–14, February 20, 2018 11

Page 13: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

more targeted approach to reinvigorating anti-tumor or anti-viral

immunity. As our data supported that these regulatory pathways

may be distinct from the exhaustion pathways mediated

by checkpoint blockade inhibitors, this suggests that these

new therapies could be used in conjunction with checkpoint

blockade to ameliorate patient outcomes.

In conclusion, our data provided mechanistic insight into the

impact of IL-10 on CD8+ T cells. We propose a model in which

IL-10 induced by infection with LCMV cl13 directly regulates

the expression ofMgat5, leading to heightened Mgat5-modified

N-glycan branching on CD8+ T cells that enhances binding of

Gal3. Elevated Gal3 binding restricts TCR:CD8 co-receptor

association and reduces TCR signaling capacity ultimately

decreasing CD8+ T cell antigen sensitivity. Thus, our data sug-

gest a regulatory pathway that leads to dysfunction of T cells

during persistent infections and identifies pathways that may

provide novel therapeutic targets to potentially enhance the

T cell response to chronic viral infections and cancer.

STAR+METHODS

Detailed methods are provided in the online version of this paper

and include the following:

d KEY RESOURCES TABLE

d CONTACT FOR REAGENT AND RESOURCE SHARING

d EXPERIMENTAL MODEL AND SUBJECT DETAILS

B Mice

B Pathogens

B Cell lines

B Primary cells

d METHOD DETAILS

B Adoptive Transfer

B Ex vivo cytokine production

B TCR signaling and Immunoblot

B FRET

B Lactose treatment

B In vivo treatments

B Branched N-glycan surface expression analysis

B PHA-L pull-down

B Cytolysis Assay

B Gene expression analysis

B CD8+ T cell transduction

B Ex vivo IL-10 treatment

B MGAT5 expression from patient samples

B ELISA

d QUANTIFICATION AND STATISTICAL ANALYSIS

d DATA AND SOFTWARE AVAILABILITY

SUPPLEMENTAL INFORMATION

Supplemental Information includes six figures and one table and can be found

with this article online at https://doi.org/10.1016/j.immuni.2018.01.006.

ACKNOWLEDGMENTS

We would like to thank Camille Stegen for flow cytometry assistance. We

would also like to thank Jeff Nolz (Oregon Health & Science University) and

members of the Richer Lab for discussion and critical comments on themanu-

script. This work was supported by startup funds from the McGill Faculty of

12 Immunity 48, 1–14, February 20, 2018

Medicine and grants from NSERC (RGPIN-2016-04713), FRQS (32807), and

CIHR (PJT-152903) to M.J.R., grants from NIH/NIAID (R01AI125446 and

R01AI127481) to N.S.B., and a grant from CIHR (MOP-126184) to C.M.K.

The Montreal hepatitis C cohort is supported by grants CIHR (MOP-133680)

and FRQS AIDS and Infectious Disease Network (Reseau SIDA-MI) to J.B.

and N.H.S. M.J.R. is supported by a New Investigator Salary Award from

CIHR and a Chercheurs-Boursiers Junior 1 salary award from FRQS. L.K.S.

is supported by a Canada Graduate Scholarship-Master’s award from CIHR.

S.M. is supported by a doctoral fellowship from the Canadian Network on

Hepatitis C (CanHepC). CanHepC is funded by a joint initiative of the CIHR

(NHC-142832) and the Public Health Agency of Canada.

AUTHOR CONTRIBUTIONS

L.K.S., G.M.B., N.S.B., J.B., N.H.S., C.M.K., andM.J.R. designed and planned

experiments. L.K.S., G.M.B., S.A.C., S.M., R.V., J.J.G., and N.S.B. performed

experiments and collected the data. L.K.S., G.M.B., S.A.C., and M.J.R.

analyzed the data. L.K.S. and M.J.R. wrote the manuscript with author input.

DECLARATION OF INTERESTS

The authors declare no competing interests.

Received: June 17, 2017

Revised: September 1, 2017

Accepted: January 2, 2018

Published: January 23, 2018

REFERENCES

Abdel-Hakeem, M.S., Boisvert, M., Bruneau, J., Soudeyns, H., and Shoukry,

N.H. (2017). Selective expansion of high functional avidity memory CD8

T cell clonotypes during hepatitis C virus reinfection and clearance. PLoS

Pathog. 13, e1006191.

Ahmed, R., Salmi, A., Butler, L.D., Chiller, J.M., and Oldstone, M.B. (1984).

Selection of genetic variants of lymphocytic choriomeningitis virus in spleens

of persistently infected mice. Role in suppression of cytotoxic T lymphocyte

response and viral persistence. J. Exp. Med. 160, 521–540.

Alexander-Miller, M.A. (2005). High-avidity CD8+ T cells: optimal soldiers in the

war against viruses and tumors. Immunol. Res. 31, 13–24.

Badr, G., Bedard, N., Abdel-Hakeem, M.S., Trautmann, L., Willems, B.,

Villeneuve, J.P., Haddad, E.K., Sekaly, R.P., Bruneau, J., and Shoukry, N.H.

(2008). Early interferon therapy for hepatitis C virus infection rescues polyfunc-

tional, long-lived CD8+ memory T cells. J. Virol. 82, 10017–10031.

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.

Bian, Y., Zhou,W., Zhao, Y., Li, X., Geng,W., Hao, R., Yang, Q., and Huang,W.

(2011). High-dose siRNAs upregulate mouse Eri-1 at both transcription and

posttranscription levels. PLoS ONE 6, e26466.

Borger, J.G., Zamoyska, R., andGakamsky, D.M. (2014). Proximity of TCR and

its CD8 coreceptor controls sensitivity of T cells. Immunol. Lett. 157, 16–22.

Brooks, D.G., Trifilo, M.J., Edelmann, K.H., Teyton, L., McGavern, D.B., and

Oldstone, M.B. (2006). Interleukin-10 determines viral clearance or persis-

tence in vivo. Nat. Med. 12, 1301–1309.

Brooks, D.G., Ha, S.J., Elsaesser, H., Sharpe, A.H., Freeman, G.J., and

Oldstone, M.B. (2008). IL-10 and PD-L1 operate through distinct pathways

to suppress T-cell activity during persistent viral infection. Proc. Natl. Acad.

Sci. USA 105, 20428–20433.

Busch, D.H., and Pamer, E.G. (1999). T cell affinity maturation by selective

expansion during infection. J. Exp. Med. 189, 701–710.

Butler, N.S., Moebius, J., Pewe, L.L., Traore, B., Doumbo, O.K., Tygrett, L.T.,

Waldschmidt, T.J., Crompton, P.D., and Harty, J.T. (2011). Therapeutic

blockade of PD-L1 and LAG-3 rapidly clears established blood-stage

Plasmodium infection. Nat. Immunol. 13, 188–195.

Page 14: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

Cawthon, A.G., and Alexander-Miller, M.A. (2002). Optimal colocalization of

TCR and CD8 as a novel mechanism for the control of functional avidity.

J. Immunol. 169, 3492–3498.

Chen, R., Belanger, S., Frederick, M.A., Li, B., Johnston, R.J., Xiao, N., Liu,

Y.C., Sharma, S., Peters, B., Rao, A., et al. (2014). In vivo RNA interference

screens identify regulators of antiviral CD4(+) and CD8(+) T cell differentiation.

Immunity 41, 325–338.

Corinti, S., Albanesi, C., la Sala, A., Pastore, S., and Girolomoni, G. (2001).

Regulatory activity of autocrine IL-10 on dendritic cell functions. J. Immunol.

166, 4312–4318.

Cui, W., Liu, Y., Weinstein, J.S., Craft, J., and Kaech, S.M. (2011). An inter-

leukin-21-interleukin-10-STAT3 pathway is critical for functional maturation

of memory CD8+ T cells. Immunity 35, 792–805.

Demetriou, M., Granovsky, M., Quaggin, S., and Dennis, J.W. (2001). Negative

regulation of T-cell activation and autoimmunity by Mgat5 N-glycosylation.

Nature 409, 733–739.

Demotte, N., Stroobant, V., Courtoy, P.J., Van Der Smissen, P., Colau, D.,

Luescher, I.F., Hivroz, C., Nicaise, J., Squifflet, J.L., Mourad, M., et al.

(2008). Restoring the association of the T cell receptor with CD8 reverses

anergy in human tumor-infiltrating lymphocytes. Immunity 28, 414–424.

Demotte, N., Wie€ers, G., Van Der Smissen, P., Moser, M., Schmidt, C.,

Thielemans, K., Squifflet, J.L., Weynand, B., Carrasco, J., Lurquin, C., et al.

(2010). A galectin-3 ligand corrects the impaired function of human CD4 and

CD8 tumor-infiltrating lymphocytes and favors tumor rejection in mice.

Cancer Res. 70, 7476–7488.

Durant, L., Watford, W.T., Ramos, H.L., Laurence, A., Vahedi, G., Wei, L.,

Takahashi, H., Sun, H.W., Kanno, Y., Powrie, F., and O’Shea, J.J. (2010).

Diverse targets of the transcription factor STAT3 contribute to T cell pathoge-

nicity and homeostasis. Immunity 32, 605–615.

Ejrnaes, M., Filippi, C.M., Martinic, M.M., Ling, E.M., Togher, L.M., Crotty, S.,

and von Herrath, M.G. (2006). Resolution of a chronic viral infection after

interleukin-10 receptor blockade. J. Exp. Med. 203, 2461–2472.

Elola, M.T., Blidner, A.G., Ferragut, F., Bracalente, C., and Rabinovich, G.A.

(2015). Assembly, organization and regulation of cell-surface receptors by

lectin-glycan complexes. Biochem. J. 469, 1–16.

Emmerich, J., Mumm, J.B., Chan, I.H., LaFace, D., Truong, H., McClanahan,

T., Gorman, D.M., and Oft, M. (2012). IL-10 directly activates and expands

tumor-resident CD8(+) T cells without de novo infiltration from secondary

lymphoid organs. Cancer Res. 72, 3570–3581.

Flynn, J.K., Dore, G.J., Hellard, M., Yeung, B., Rawlinson, W.D., White, P.A.,

Kaldor, J.M., Lloyd, A.R., and Ffrench, R.A.; ATAHC Study Group (2011).

Early IL-10 predominant responses are associated with progression to chronic

hepatitis C virus infection in injecting drug users. J. Viral Hepat. 18, 549–561.

Frebel, H., Richter, K., and Oxenius, A. (2010). How chronic viral infections

impact on antigen-specific T-cell responses. Eur. J. Immunol. 40, 654–663.

Grebely, J., Morris, M.D., Rice, T.M., Bruneau, J., Cox, A.L., Kim, A.Y.,

McGovern, B.H., Shoukry, N.H., Lauer, G., Maher, L., et al.; InC Study

Group (2013). Cohort profile: the international collaboration of incident HIV

and hepatitis C in injecting cohorts (InC3) study. Int. J. Epidemiol. 42,

1649–1659.

Grigorian, A., and Demetriou, M. (2011). Mgat5 deficiency in T cells and exper-

imental autoimmune encephalomyelitis. ISRN Neurol. 2011, 374314.

Ilarregui, J.M., Bianco, G.A., Toscano, M.A., and Rabinovich, G.A. (2005). The

coming of age of galectins as immunomodulatory agents: impact of these

carbohydrate binding proteins in T cell physiology and chronic inflammatory

disorders. Ann. Rheum. Dis. 64 (Suppl 4 ), iv96–iv103.

Ip, W.K.E., Hoshi, N., Shouval, D.S., Snapper, S., and Medzhitov, R. (2017).

Anti-inflammatory effect of IL-10 mediated by metabolic reprogramming of

macrophages. Science 356, 513–519.

Iwamaru, A., Szymanski, S., Iwado, E., Aoki, H., Yokoyama, T., Fokt, I., Hess,

K., Conrad, C., Madden, T., Sawaya, R., et al. (2007). A novel inhibitor of the

STAT3 pathway induces apoptosis in malignant glioma cells both in vitro

and in vivo. Oncogene 26, 2435–2444.

Kang, R., Saito, H., Ihara, Y., Miyoshi, E., Koyama, N., Sheng, Y., and

Taniguchi, N. (1996). Transcriptional regulation of the N-acetylglucosaminyl-

transferase V gene in human bile duct carcinoma cells (HuCC-T1) is mediated

by Ets-1. J. Biol. Chem. 271, 26706–26712.

Kobayashi, F., Morii, T., Matsui, T., Fujino, T., Watanabe, Y., Weidanz, W.P.,

and Tsuji, M. (1996). Production of interleukin 10 during malaria caused by le-

thal and nonlethal variants of Plasmodium yoelii yoelii. Parasitol. Res. 82,

385–391.

Krawczyk, C.M., Holowka, T., Sun, J., Blagih, J., Amiel, E., DeBerardinis, R.J.,

Cross, J.R., Jung, E., Thompson, C.B., Jones, R.G., and Pearce, E.J. (2010).

Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic

cell activation. Blood 115, 4742–4749.

Kuball, J., Hauptrock, B., Malina, V., Antunes, E., Voss, R.H., Wolfl, M., Strong,

R., Theobald, M., and Greenberg, P.D. (2009). Increasing functional avidity of

TCR-redirected T cells by removing defined N-glycosylation sites in the TCR

constant domain. J. Exp. Med. 206, 463–475.

Kwon, D.S., and Kaufmann, D.E. (2010). Protective and detrimental roles of

IL-10 in HIV pathogenesis. Eur. Cytokine Netw. 21, 208–214.

Livak, K.J., and Schmittgen, T.D. (2001). Analysis of relative gene expression

data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method.

Methods 25, 402–408.

Malherbe, L., Hausl, C., Teyton, L., and McHeyzer-Williams, M. (2004). Clonal

selection of helper T cells is determined by an affinity threshold with no further

skewing of TCR binding properties. Immunity 21, 669–679.

Mannino, M.H., Zhu, Z., Xiao, H., Bai, Q., Wakefield, M.R., and Fang, Y. (2015).

The paradoxical role of IL-10 in immunity and cancer. Cancer Lett. 367,

103–107.

Moore, K.W., de Waal Malefyt, R., Coffman, R.L., and O’Garra, A. (2001).

Interleukin-10 and the interleukin-10 receptor. Annu. Rev. Immunol. 19,

683–765.

Ng, C.T., and Oldstone, M.B. (2014a). IL-10: achieving balance during persis-

tent viral infection. Curr. Top. Microbiol. Immunol. 380, 129–144.

Ng, C.T., and Oldstone, M.B.A. (2014b). IL-10: achieving balance during

persistent viral infection. In Interleukin-10 in Health and Disease, S. Fillatreau

and A. Ogarra, eds. (Berlin: Springer-Verlag Berlin), pp. 129–144.

Paddison, P.J., Cleary, M., Silva, J.M., Chang, K., Sheth, N., Sachidanandam,

R., and Hannon, G.J. (2004). Cloning of short hairpin RNAs for gene knock-

down in mammalian cells. Nat. Methods 1, 163–167.

Parish, I.A., Marshall, H.D., Staron, M.M., Lang, P.A., Br€ustle, A., Chen, J.H.,

Cui, W., Tsui, Y.C., Perry, C., Laidlaw, B.J., et al. (2014). Chronic viral infection

promotes sustained Th1-derived immunoregulatory IL-10 via BLIMP-1. J. Clin.

Invest. 124, 3455–3468.

Perica, K., Bieler, J.G., Edidin, M., and Schneck, J. (2012). Modulation of MHC

binding by lateral association of TCR and coreceptor. Biophys. J. 103,

1890–1898.

Pircher, H., Michalopoulos, E.E., Iwamoto, A., Ohashi, P.S., Baenziger, J.,

Hengartner, H., Zinkernagel, R.M., and Mak, T.W. (1987). Molecular analysis

of the antigen receptor of virus-specific cytotoxic T cells and identification of

a new V alpha family. Eur. J. Immunol. 17, 1843–1846.

Richer, M.J., Nolz, J.C., and Harty, J.T. (2013). Pathogen-specific inflamma-

tory milieux tune the antigen sensitivity of CD8(+) T cells by enhancing T cell

receptor signaling. Immunity 38, 140–152.

Santin, A.D., Hermonat, P.L., Ravaggi, A., Bellone, S., Pecorelli, S., Roman,

J.J., Parham, G.P., and Cannon, M.J. (2000). Interleukin-10 increases Th1

cytokine production and cytotoxic potential in human papillomavirus-specific

CD8(+) cytotoxic T lymphocytes. J. Virol. 74, 4729–4737.

Slifka, M.K., and Whitton, J.L. (2001). Functional avidity maturation of CD8(+)

T cells without selection of higher affinity TCR. Nat. Immunol. 2, 711–717.

Smith-Garvin, J.E., Koretzky, G.A., and Jordan, M.S. (2009). T cell activation.

Annu. Rev. Immunol. 27, 591–619.

Sun, Z., Fourcade, J., Pagliano, O., Chauvin, J.M., Sander, C., Kirkwood, J.M.,

and Zarour, H.M. (2015). IL10 and PD-1 cooperate to limit the activity of tumor-

specific CD8+ T cells. Cancer Res. 75, 1635–1644.

Immunity 48, 1–14, February 20, 2018 13

Page 15: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

Vigano, S., Utzschneider, D.T., Perreau, M., Pantaleo, G., Zehn, D., and Harari,

A. (2012). Functional avidity: a measure to predict the efficacy of effector

T cells? Clin. Dev. Immunol. 2012, 153863.

Walker, L.J., Sewell, A.K., and Klenerman, P. (2010). T cell sensitivity and the

outcome of viral infection. Clin. Exp. Immunol. 159, 245–255.

Wherry, E.J., and Kurachi, M. (2015). Molecular and cellular insights into T cell

exhaustion. Nat. Rev. Immunol. 15, 486–499.

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

14 Immunity 48, 1–14, February 20, 2018

distribution and results in distinct stages of functional impairment. J. Virol.

77, 4911–4927.

Wilson, E.B., and Brooks, D.G. (2011). The role of IL-10 in regulating immunity

to persistent viral infections. Curr. Top. Microbiol. Immunol. 350, 39–65.

Yi, J.S., Cox, M.A., and Zajac, A.J. (2010). T-cell exhaustion: characteristics,

causes and conversion. Immunology 129, 474–481.

Zehn, D., Lee, S.Y., and Bevan, M.J. (2009). Complete but curtailed T-cell

response to very low-affinity antigen. Nature 458, 211–214.

Page 16: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

STAR+METHODS

KEY RESOURCES TABLE

REAGENT or RESOURCE SOURCE IDENTIFIER

Antibodies

Anti-mouse Akt NEB Cell Signaling Cat#9272, RRID:AB_329827

Anti-mouse Akt (phospho) NEB Cell Signaling Cat#9275, RRID:AB_329828

Anti-mouse CD210 (blocking) BioXcell Cat#BE0050, RRID:AB_1107611

Anti-mouse CD28 BioLegend Cat#102111, RRID:AB_312876

Anti-mouse CD28 - APC BioLegend Cat#102109, RRID:AB_312874

Anti-mouse CD3ε BioLegend Cat#100331, RRID:AB_1877073

Anti-mouse CD3ε - biotin BioLegend Cat#100303, RRID:AB_312668

Anti-mouse CD3ε - PE BioLegend Cat#100307, RRID:AB_312672

Anti-mouse CD8a - PacificBlue BioLegend Cat#100725, RRID:AB_493425

Anti-mouse CD8a - APC BioLegend Cat#100712, RRID:AB_312751

Anti-mouse CD8a - FITC BioLegend Cat#100706, RRID:AB_312745

Anti-mouse CTLA4 - PE BioLegend Cat#106305, RRID:AB_313254

Anti-mouse ERK1/2 NEB Cell Signaling Cat#9102, RRID:AB_330744

Anti-mouse ERK1/2 (phospho) NEB Cell Signaling Cat#9101, RRID:AB_331646

Anti-mouse Galectin 3 - PE BioLegend Cat#125406, RRID:AB_2136762

Anti-mouse IFNg - FITC BioLegend Cat#505806, RRID:AB_315400

Anti-mouse IFNg - APC BioLegend Cat#505810, RRID:AB_315404

Anti-mouse Lag3 - PE BioLegend Cat#125207, RRID:AB_2133344

Anti-mouse PD-1 - PE-Cy7 BioLegend Cat#135216, RRID:AB_10689635

Anti-mouse PLCg1 NEB Cell Signaling Cat#2822, RRID:AB_2163702

Anti-mouse PLCg1 (phospho) NEB Cell Signaling Cat#2821, RRID:AB_33085

Anti-mouse TCRa/b - biotin Cedarlane Cat#CL7200B, RRID:AB_10086441

Anti-mouse TCR Va2 - PE BioLegend Cat#127808, RRID:AB_1134183

Anti-mouse TCR Vb8.1 - FITC eBioscience Cat#11-5813-81, RRID:AB_465261

Anti-mouse Thy1.1 - PerCP-Cy5.5 BioLegend Cat#202516, RRID:AB_961437

Anti-mouse Thy1.1 - PE BioLegend Cat#202524, RRID:AB_1595524

Anti-mouse Thy1.1 - FITC BioLegend Cat#202503, RRID:AB_314014

Anti-mouse Thy1.2 - PerCP-Cy5.5 BioLegend Cat#140321, RRID:AB_2562695

Anti-mouse Thy1.2 - PE BioLegend Cat#140307, RRID:AB_10643585

Anti-mouse Thy1.2 - FITC BioLegend Cat#140304, RRID:AB_10642812

Anti-mouse Tim3 - PE BioLegend Cat#134003, RRID:AB_1626181

Anti-mouse TNFa - PE BioLegend Cat#506305, RRID:AB_315426

Anti-mouse ZAP-70 NEB Cell Signaling Cat#3165, RRID:AB_2218656

Anti-mouse ZAP-70 (phospho) NEB Cell Signaling Cat#2701, RRID:AB_331600

Bacterial and Viral Strains

E. coli DH5a Dr. Connie Krawczyk Lab TaxID: 668389

LCMV Armstrong Dr. John Harty Lab N/A

LCMV clone 13 Dr. Tania Watts Lab N/A

Chemicals, Peptides, and Recombinant Proteins

Recombinant mouse Galectin 3 BioLegend Cat#599804

Phaseolus vulgaris Leukagglutenin (PHA-L) - biotin Cedarlane Cat#BA-1801-2

Agarose bound PHA-L Vector Labs Cat#AL-1113

Streptavidin BioLegend Cat#280302

Streptavidin - PE BioLegend Cat#405203

(Continued on next page)

Immunity 48, 1–14.e1–e5, February 20, 2018 e1

Page 17: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

Continued

REAGENT or RESOURCE SOURCE IDENTIFIER

Streptavidin-HRP BioLegend Cat#405210

Brefeldin A BioLegend Cat#420601

IC Fixation buffer eBioscience Cat#00-8222-49

10x Permbuffer eBioscience Cat#00-8333-56

GP 33-41 BioSyn KAVYNFATM

Lactose Fisher Cat#L5500

D-(+)-Glucose Sigma Cat#G-8270

efluor 450 Proliferation Dye eBioscience Cat#65-0842-85

Critical Commercial Assays

Mouse Galectin 3 ELISA kit ThermoFisher Scientific Cat#EMLGALS3

Experimental Models: Cell Lines

EL4 Dr. John Harty Lab ATCC# TIB-39, RRID:CVCL_0255

Vero Dr. Steve Varga Lab ATCC# CCL-81, RRID:CVCL_0059

HEK293T Dr. Connie Krawczyk Lab ATCC# CRL-3216, RRID:CVCL_0063

Experimental Models: Organisms

C57BL6/J mice Charles River Cat#27

B6.129P2(B6)-Il10tm1Cgn/J Jackson Laboratories Cat#2250

B6.Cg-Lgals3tmPoi/J Jackson Laboratories Cat#6338

Oligonucleotides

Primer sequences Table S1 N/A

Recombinant DNA

pLMPd-Ametrine N/A Chen et al., 2014

pCL-Eco packaging vector Addgene Cat# 21371

Software and Algorithms

Flowjo 9.9 Tree Star N/A

Prism 7 GraphPad N/A

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

CONTACT FOR REAGENT AND RESOURCE SHARING

Further information and requests for reagents should be directed to the Lead Contact, Martin J. Richer ([email protected])

EXPERIMENTAL MODEL AND SUBJECT DETAILS

MiceC57BL/6 and Il10�/� mice were originally purchased from Charles River Laboratories and The Jackson Laboratory, respectively

and bred in house. Mice with P14 TCR-tg CD8+ T cells were previously described (Pircher et al., 1987) and were provided by

Dr. A. Lamarre (INRS-Institut Armand-Frappier) and bred in house. Lgals3�/�mice were provided by Dr. D. Sheppard (McGill Univer-

sity). Infected mice where housed in biocontainment level 2 and all animal procedures were carried out in accordance with the

Canadian Council on Animal Care and were approved by the McGill University Animal Care Committee. 6-12 week old mice of

both sexes were used for all experiments

PathogensLCMV Armstrong and LCMV clone 13 were kindly provided by Dr. J. Harty (University of Iowa) and Dr. T. Watts (University of Toronto)

from a strain originally propagated by Dr. M. Oldstone (The Scripps Research Institute), respectively. LCMV was propagated as

described (Slifka and Whitton, 2001). Mice were infected with 2 3 105 plaque forming units (PFU) of LCMV Armstrong by intraper-

itoneal (i.p.) injection, or 2 3 106 PFU LCMV clone 13 intravenous (i.v.) injection, respectively (Richer et al., 2013; Wherry et al.,

2003). Viral titers were determined by plaque assay conducted on Vero cells (Ahmed et al., 1984). Briefly, Vero cell monolayers

were infected with 100 mL of serially diluted kidney or liver sample and incubated for 90 minutes at 37�C in 5% CO2. Agarose overlay

was added to infected cells and placed in incubator for 3 days at 37�C in 5% CO2. Cells were then stained with agarose overlay

supplemented with 1% neutral red for 2 days at 37�C in 5% CO2 and plaques were counted.

e2 Immunity 48, 1–14.e1–e5, February 20, 2018

Page 18: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

Plasmodium yoelii yoelii clone 17XNL expressing GP33-41 of LCMVwas routinely passaged throughmosquitoes. 13 104 naive P14

TCR-tg CD8+ T cells were transferred into congenically mismatched recipients at day�1. 24 hours later mice received 13 106 para-

sitized red blood cells by i.v. serial transfer to initiate infections. At day 7 post-infectionmicewere sacrificed and P14 cells were sorted

by flow cytometry.

Cell linesAll cells were cultured at 37�C with 5% CO2. Green monkey VERO cells (female origin, provided by Dr. S. Varga, University of Iowa)

and Human embryonic kidney tubule HEK293T cells (female origin) were cultured in Dulbecco’s Modified Eagle Medium supple-

mented with 1% non-essential amino acids, 1% penicillin and streptomycin, 2mM L-glutamine, 1% sodium pyruvate and 10%

heat-inactivated FBS. EL4 (female origin, provided by Dr. J. Harty, University of Iowa) suspension cells were cultured in RPMI

1640 supplemented with 1% penicillin and streptomycin, 2 nM L-glutamine, 25 mM HEPES, 50 mM b-mercaptoethanol and 10%

heat-inactivated FBS.

Primary cellsAll cells were cultured at 37�C with 5% CO2. For routine ex vivo peptide stimulation total splenocytes were cultured in RPMI 1640

supplemented with 1% penicillin and streptomycin, 2 nM L-glutamine, 25 mM HEPES, 50 mM b-mercaptoethanol and 10% heat-in-

activated FBS. For ex vivo activation, expansion and transduction of P14 TCR-tg and wild-type CD8+ T cells, cells were cultured in

Iscove’s Modified Dulbecco’s Medium supplemented with 1% penicillin and streptomycin, 2 nM L-glutamine, 25 mMHEPES, 50 mM

b-mercaptoethanol and 10% heat-inactivated FBS.

METHOD DETAILS

Adoptive Transfer2 – 100 3 103 naive P14 TCR-tg CD8+ T cells were transferred by i.v. injection into congenically mismatched recipients at the Thy1

locus. 1 day following transfer mice were infected with the appropriate strain of LCMV. At day 8 post-infection mice were sacrificed

and splenic CD8+ T cells were analyzed.

Ex vivo cytokine production1 – 3 3 106 splenocytes were incubated ex vivo with titrated concentrations of GP33-41 peptide in the presence of BFA. Cells were

stimulated for 6 hours at 37�C, 5% CO2 and then stained for the production of cytokines by intracellular cytokine staining.

TCR signaling and Immunoblot1 - 10 3 104 naive P14 TCR-tg CD8+ T cells were injected i.v. into naive congenically mismatched recipients; 1 day later mice were

infected with LCMV Arm or LCMV cl13, as indicated. On day 8 post-infection, spleens were harvested and transgenic cells were iso-

lated by Thy1.1 or Thy1.2-PE positive-selection, accordingly. In brief, cells were stained with the appropriate PE conjugated antibody

and purified using anti-PE-magnetic separation according to standard AutoMACS protocols (Miltenyi Biotec). 1 3 107 cells were

stimulated by CD3ε crosslinking or PMA stimulation at 37�C; following stimulation cells were lysed with 25 – 50 mL of NP-40 lysis

buffer, accordingly. 15 – 25 mg of protein was resolved by SDS-PAGE, transferred to PVDFmembranes and probedwith the indicated

antibodies. Antibody binding was detected using goat-anti-rabbit conjugated to horseradish peroxidase and Amersham Prime ECL.

FRETTCR-CD8 FRET was measured using flow cytometry. 1 – 5 3 106 total splenocytes were incubated for 30 minutes with PE-conju-

gated anti-Va2 (a component of the transgenic TCR) as a fluorescence donor and APC-conjugated anti-CD8a as a fluorescence

acceptor. Samples were stained with either antibody (EPE, or EAPC), both (EBoth) or neither (Enone). Cells were concurrently labeled

with CD3ε-biotin and stimulated by crosslinking with streptavidin for 2 minutes. Stimulated samples were subsequently fixed with

IC Fixation buffer for 10 minutes and quantified by flow cytometry. FRET emission was assessed by flow cytometry without compen-

sation, observing emission in the APC wavelength without direct laser excitation. FRET efficiency was calculated in FRET units

(Perica et al., 2012).

FRET unit = (E3Both – E3none) – [(E3APC -E3none)x(E2both/E2APC)] – [(E3PE-E3none)x(E1Both/E1PE)]

E1: emission in the donor channel upon excitation of the donor

E2: emission in the acceptor channel upon excitation of the acceptor

E3: emission in the acceptor channel upon excitation of the donor

Lactose treatmentFor experiments measuring antigen sensitivity: 1 – 33 106 splenocytes were incubated with titrated concentrations of peptide in the

presence of BFA as described above. Prior to the 6 hour incubation cells were supplemented with 50 mM D-Lactose. For signaling

Immunity 48, 1–14.e1–e5, February 20, 2018 e3

Page 19: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

experiments and FRET analysis: purified or total cells were supplemented with 50 mM D-Lactose and incubated for 30 min in 37�Cwater bath to disrupt galectin binding (Demetriou et al., 2001). Cells were then processed as described above.

In vivo treatmentsFor experiments using anti-CD210 blocking antibody: mice received 50,000 congenically marked P14 TCR-tg CD8+ T cells by adop-

tive transfer, the following day mice were infected with LCMV cl13, as described above. At days 1 and 5 post-infection mice were

treated with 500 mg of rat-anti-CD210 or rat-IgG1 isotype control by i.p. injection. Mice were sacrificed and analyzed at day 8

post-infection.

Branched N-glycan surface expression analysisCells were treated with 50 mMD-Lactose to remove bound galectins, which may impair staining through steric hindrance. Cells were

stained with fluorescently labeled antibodies for 20minutes, then washed and fixed. Cells were incubated with 50 mg/mL biotinylated

Phaseolus vulgaris Leukoagglutinin (PHA-L) for 1+ hour at room temperature, washed and incubatedwith PE-conjugated streptavidin

and analyzed by flow cytometry.

PHA-L pull-downP14 cells were enriched from total splenocytes by standard AutoMACS protocol as described above. Isolated cells were treated with

50 mM D-Lactose for 30 minutes at 37�C, to remove any surface bound galectins and expose branched N-glycans. Cells were then

pelleted by centrifugation, lysed in NP-40 lysis buffer, and quantified using a Bradford assay. 10% of whole cell lysate was reserved

and loaded as the input fraction. 250 mg of total protein was incubated with 150 mg of PHA-L conjugated agarose beads at room tem-

perature, overnight, with agitation. Beads were pelleted by centrifugation and washed 4 times with an equal volume of lysis buffer.

Proteins were eluted using Laemmli sample buffer (Sigma Aldrich), and detected by SDS-PAGE followed by immunoblotting with

anti-TCRa/b.

Cytolysis AssayEL4 cells were labeled with efluor 450 proliferation dye at either 100 nM or 0.05 nM (e450hi and e450lo, respectively). e450lo cells were

then incubated for 1 hour at 37�C with 5% CO2 in the presence of 0.05 nM GP33-41 peptide. Following incubation, both cell popula-

tions were washed and counted. e450hi cells were mixed at a 1:1 ratio with e450lo-target cells. 13 106 total EL4s were then seeded

into a 96 well plate in 100 mL of medium. P14 TCR-tg CD8+ T cells were enriched from the spleen of day 8 infected mice according to

standard Automacs protocols. 1.253 105 enriched P14 cells/well were co-cultured with EL4s (a 1:4:4 ratio of effector CD8+ T cells:

e450lo GP33-41 pulsed target cells: e450hi non-target cells) and incubated at 37�Cwith 5%CO2 for 2 hours. Following incubation, cells

were fixed with IC fixation buffer and analyzed by flow cytometry. Specific lysis was determined by the proportion of e450lo target

cells to e450hi non-targets, relative to EL4s cultured alone.

Specific Lysis = 100% - ([(%e450lo target cells co-cultured /%e450hi non-target cells co-cultured) / (%e450lo target cells alone /%

e450hi non-target cells alone)] x 100%)

Gene expression analysisFor RT-qPCR analysis, P14 cells were isolated by PE-selection as described above. RNAwas then extracted (Trizol reagent) and 1 mg

of total RNA was used to generate cDNA with iScript reverse transcriptase (Bio-Rad). RT-PCR analysis was then conducted using

SensiFAST SYBR (Bioline). Transcript expression was normalized to TATA binding protein as an internal control and depicted as a

relative fold change using the DDCt method, compared to the mean of the control group (Livak and Schmittgen, 2001).

CD8+ T cell transductionNaive P14 cells were isolated from spleens by negative CD8 selection (STEMCELL technologies 19853). 23 106 cells per well were

stimulated with plate bound CD3ε and CD28 in the presence of 20 units/mL of recombinant murine IL-2 (eBioscience). 18 hours later,

activated cells were either transduced using retrovirus produced in HEK293Ts transfected with LMPd-based retroviral vectors pro-

vided by Dr. M. Pipkin (The Scripps Research Institute, Florida) and Dr. S. Crotty (La Jolla Institute for Allergy and Immunology) en-

coding the shRNA of interest (Chen et al., 2014; Paddison et al., 2004) or mock-transduced. Cells were allowed to recover for 6 hours

prior to adoptive transfer into recipient mice that were infected with LCMV 6 hours prior.

Ex vivo IL-10 treatmentNaive CD8+ T cells were isolated from spleens by negative CD8 selection (STEMCELL Technologies). 23 106 cells/mLwere activated

with plate bound CD3ε/CD28 in the presence of 20 units/mL of IL-2. Cells were treated with or without various doses of recombinant

murine IL-10 (eBioscience) and with or without 0.5 nmol/mL the STAT3 inhibitor WP1066 (Sigma 573097). After 2 days, cells were

transferred to plates without CD3ε/CD28 and maintained up to day 8 with or without addition of recombinant murine IL-10 as indi-

cated at 37�C with 5% CO2.

e4 Immunity 48, 1–14.e1–e5, February 20, 2018

Page 20: Interleukin-10 Directly Inhibits CD8+ T Cell Function by ... · Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity

Please cite this article in press as: Smith et al., Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to DecreaseAntigen Sensitivity, Immunity (2018), https://doi.org/10.1016/j.immuni.2018.01.006

MGAT5 expression from patient samplesStudy subjects were enrolled among people who inject drugs (PWIDs) participating in the Montreal Acute Hepatitis C Cohort Study

(HEPCO) (Grebely et al., 2013). This study was approved by the Institutional Ethics Committee of CRCHUM (Protocol SL05.014). All

samples were anonymized. Chronic HCV infection was identified in participants who tested positive for HCV RNA for more than

6 months post initial infection as previously described (Badr et al., 2008; Grebely et al., 2013). CD8+ T cells were isolated from cry-

opreserved peripheral blood mononuclear cells (PBMCS) from five chronically infected or HCV naive participants by MACS separa-

tion (Miltenyi Biotech). RNA was isolated using RNeasy Plus kit (QIAGEN). cDNA synthesis and RT-qPCR were performed as

described above.

ELISAGalectin 3 abundance in the serum was determined by sandwich ELISA. Serum samples were diluted 1:4, 1:8 and 1:16 in assay

diluent and incubated overnight at 4�C. Secondary anti-galectin-biotin was added for 1 hour at room temperature, streptavidin-

HRP was added for 45 minutes at room temperature. Samples were developed using TMB substrate and read by measuring absor-

bance at 450 nm and 550 nm. Galectin 3 was quantified by comparison relative to serially diluted galectin 3 protein standard.

QUANTIFICATION AND STATISTICAL ANALYSIS

Data were analyzed with GraphPad Prism software. The Specific tests used to determine statistical significance are indicated in each

figure legend. P values of less than 0.05 were considered statistically significant.

DATA AND SOFTWARE AVAILABILITY

Data were analyzed using Flowjo andGraphPad Prism analysis software. Both packages are publically available through commercial

vendors.

Immunity 48, 1–14.e1–e5, February 20, 2018 e5