immune regulation copyright © 2017 interleukin-10 from cd4 ...€¦ · laidlaw et al., sci....

11
Laidlaw et al., Sci. Immunol. 2, eaan4767 (2017) 20 October 2017 SCIENCE IMMUNOLOGY | RESEARCH ARTICLE 1 of 10 IMMUNE REGULATION Interleukin-10 from CD4 + follicular regulatory T cells promotes the germinal center response Brian J. Laidlaw, 1 * Yisi Lu, 1 * Robert A. Amezquita, 1 Jason S. Weinstein, 2 Jason A. Vander Heiden, 3 Namita T. Gupta, 3 Steven H. Kleinstein, 1,3,4 Susan M. Kaech, 1 Joe Craft 1,2‡ CD4 + follicular regulatory T (T fr ) cells suppress B cell responses through modulation of follicular helper T (T fh ) cells and germinal center (GC) development. We found that T fr cells can also promote the GC response through provision of interleukin-10 (IL-10) after acute infection with lymphocytic choriomeningitis virus (LCMV). Sensing of IL-10 by B cells was necessary for optimal development of the GC response. GC B cells formed in the absence of T reg cell– derived IL-10 displayed an altered dark zone state and decreased expression of the transcription factor Forkhead box protein 1 (FOXO1). IL-10 promoted nuclear translocation of FOXO1 in activated B cells. These data indicate that T fr cells play a multifaceted role in the fine-tuning of the GC response and identify IL-10 as an important mediator by which T fr cells support the GC reaction. INTRODUCTION The germinal center (GC) response is essential for the production of memory B cells and class-switched, long-lived plasma cells that pro- duce high-affinity antibodies (1). Understanding the mechanisms regulating the GC response is of interest because of the potential to harness this knowledge to bolster or, in the cases of autoimmunity and B cell lymphomas, restrain the GC reaction (2). Follicular helper T (T fh ) cells are a specialized subset of effector CD4 + T cells that ex- press the transcription factors Bcl6 and Ascl2, and the B cell follicle- homing chemokine receptor CXCR5, and reside within the GC (3, 4). T fh cells regulate the GC response through secretion of cytokines [e.g., interleukin-21 (IL-21), IL-4, and interferon- (IFN-)] and expres- sion of surface ligands such as CD40L that signal to GC B cells and promote their maturation (5). Competition for T fh cell help regulates B cell selection within the GC because high-affinity B cells preferen- tially interact with T fh cells and receive signals promoting the further proliferation and somatic hypermutation of their immunoglobulin (Ig) genes (6, 7). A subset of effector Foxp3 + regulatory CD4 + T (T reg ) cells that ex- press CXCR5 and Bcl6 was recently described (810). These cells, known as follicular regulatory T (T fr ) cells, originate from thymic-derived Foxp3 + cells or naïve cells and reside within the follicles and GC in mice and humans, where they serve to modulate the magnitude and quality of the GC and T fh cell responses (813). T fr cells express the inhibitory co-receptor CTLA4, which is essential for their restraint of the GC response (14, 15). CTLA4 suppresses the latter response by modulating B cell expression of B7-2 (CD86) outside GCs (15) or by acting on GC B cells, either dependently or independently of B7-1 (CD80) and B7-2 (1416). It may also function to control T fh cell gen- eration directly by altering CD28 engagement (17). Although T fr cells deficient in CTLA4 have impaired suppressive ability in vivo, it is likely that both T fr cells and follicular nonresident T reg cells act through CTLA4 to restrain the GC response (14, 15). T fr cells may also modulate the GC response through pathways independent of CTLA4. T reg cells regulate immune cells through con- trol of IL-2 availability, surface expression of ectoenzymes (CD39 and CD73), inhibitory receptors (e.g., CTLA4 and Lag3), and secretion of cytokines such as IL-10, IL-35, and TGF- (transforming growth factor–) (18). During influenza infection, T reg cells indirectly pro- mote T fh cell differentiation and, subsequently, the GC response by limiting T cell exposure to IL-2 (19). IL-2 signaling through signal transducer and activator of transcription 5 (STAT5) and via Akt and mechanistic target of rapamycin (mTOR) can impair T fh cell differ- entiation by suppressing Bcl6 expression and up-regulating that of Blimp1 (20). Whether T fr cells may directly regulate the GC response through similar mechanisms remains ill defined. T reg cell production of IL-10 is essential for the regulation of in- flammation at environmental interfaces, such as the colon and lung, and can enhance the development of functional memory CD8 + T cells during acute lymphocytic choriomeningitis virus (LCMV) infection through suppression of inflammation in the spleen (2123). IL-10 can also promote B cell proliferation, survival, and differentiation into antibody-secreting plasma cells (2428), although it is not known whether T fr cell–derived IL-10 can modulate the GC response. Here, we demonstrate that it does modulate the GC response during acute viral infection. These findings reveal that T fr cells play a multifaceted role in the fine-tuning of the GC response and are capable of promot- ing or suppressing the GC response depending on the signals provided and the context in which these signals are received. RESULTS T reg cell–derived IL-10 promotes B cell differentiation and GC development To examine IL-10 production by T fr cells, we infected 10BiT IL-10 re- porter mice acutely with the Armstrong strain of LCMV and assessed T fr cell phenotype and kinetics. These reporter mice have a bacterial artificial chromosome transgene containing the Il10 gene locus, with the Thy1.1 complementary DNA (containing a stop codon) replacing the endogenous coding segment of exon 1 of the Il10 locus such that 1 Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA. 2 Department of Internal Medicine (Rheumatology), Yale University School of Medicine, New Haven, CT 06520, USA. 3 Interdepartmental Program in Computational Biology and Bioinformatics, Yale University School of Medicine, New Haven, CT 06520, USA. 4 Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA. *These authors contributed equally to this work. †Present address: Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA. ‡Corresponding author. Email: [email protected] Copyright © 2017 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works by guest on September 25, 2020 http://immunology.sciencemag.org/ Downloaded from

Upload: others

Post on 25-Jul-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: IMMUNE REGULATION Copyright © 2017 Interleukin-10 from CD4 ...€¦ · Laidlaw et al., Sci. Immunol. 2, eaan4767 2017 20 October 2017 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 10

Laidlaw et al., Sci. Immunol. 2, eaan4767 (2017) 20 October 2017

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

1 of 10

I M M U N E R E G U L A T I O N

Interleukin-10 from CD4+ follicular regulatory T cells promotes the germinal center responseBrian J. Laidlaw,1*† Yisi Lu,1* Robert A. Amezquita,1 Jason S. Weinstein,2 Jason A. Vander Heiden,3 Namita T. Gupta,3 Steven H. Kleinstein,1,3,4 Susan M. Kaech,1 Joe Craft1,2‡

CD4+ follicular regulatory T (Tfr) cells suppress B cell responses through modulation of follicular helper T (Tfh) cells and germinal center (GC) development. We found that Tfr cells can also promote the GC response through provision of interleukin-10 (IL-10) after acute infection with lymphocytic choriomeningitis virus (LCMV). Sensing of IL-10 by B cells was necessary for optimal development of the GC response. GC B cells formed in the absence of Treg cell–derived IL-10 displayed an altered dark zone state and decreased expression of the transcription factor Forkhead box protein 1 (FOXO1). IL-10 promoted nuclear translocation of FOXO1 in activated B cells. These data indicate that Tfr cells play a multifaceted role in the fine-tuning of the GC response and identify IL-10 as an important mediator by which Tfr cells support the GC reaction.

INTRODUCTIONThe germinal center (GC) response is essential for the production of memory B cells and class-switched, long-lived plasma cells that pro-duce high-affinity antibodies (1). Understanding the mechanisms regulating the GC response is of interest because of the potential to harness this knowledge to bolster or, in the cases of autoimmunity and B cell lymphomas, restrain the GC reaction (2). Follicular helper T (Tfh) cells are a specialized subset of effector CD4+ T cells that ex-press the transcription factors Bcl6 and Ascl2, and the B cell follicle- homing chemokine receptor CXCR5, and reside within the GC (3, 4). Tfh cells regulate the GC response through secretion of cytokines [e.g., interleukin-21 (IL-21), IL-4, and interferon- (IFN-)] and expres-sion of surface ligands such as CD40L that signal to GC B cells and promote their maturation (5). Competition for Tfh cell help regulates B cell selection within the GC because high-affinity B cells preferen-tially interact with Tfh cells and receive signals promoting the further proliferation and somatic hypermutation of their immunoglobulin (Ig) genes (6, 7).

A subset of effector Foxp3+ regulatory CD4+ T (Treg) cells that ex-press CXCR5 and Bcl6 was recently described (8–10). These cells, known as follicular regulatory T (Tfr) cells, originate from thymic-derived Foxp3+ cells or naïve cells and reside within the follicles and GC in mice and humans, where they serve to modulate the magnitude and quality of the GC and Tfh cell responses (8–13). Tfr cells express the inhibitory co-receptor CTLA4, which is essential for their restraint of the GC response (14, 15). CTLA4 suppresses the latter response by modulating B cell expression of B7-2 (CD86) outside GCs (15) or by acting on GC B cells, either dependently or independently of B7-1 (CD80) and B7-2 (14–16). It may also function to control Tfh cell gen-eration directly by altering CD28 engagement (17). Although Tfr cells

deficient in CTLA4 have impaired suppressive ability in vivo, it is likely that both Tfr cells and follicular nonresident Treg cells act through CTLA4 to restrain the GC response (14, 15).

Tfr cells may also modulate the GC response through pathways independent of CTLA4. Treg cells regulate immune cells through con-trol of IL-2 availability, surface expression of ectoenzymes (CD39 and CD73), inhibitory receptors (e.g., CTLA4 and Lag3), and secretion of cytokines such as IL-10, IL-35, and TGF- (transforming growth factor–) (18). During influenza infection, Treg cells indirectly pro-mote Tfh cell differentiation and, subsequently, the GC response by limiting T cell exposure to IL-2 (19). IL-2 signaling through signal transducer and activator of transcription 5 (STAT5) and via Akt and mechanistic target of rapamycin (mTOR) can impair Tfh cell differ-entiation by suppressing Bcl6 expression and up-regulating that of Blimp1 (20). Whether Tfr cells may directly regulate the GC response through similar mechanisms remains ill defined.

Treg cell production of IL-10 is essential for the regulation of in-flammation at environmental interfaces, such as the colon and lung, and can enhance the development of functional memory CD8+ T cells during acute lymphocytic choriomeningitis virus (LCMV) infection through suppression of inflammation in the spleen (21–23). IL-10 can also promote B cell proliferation, survival, and differentiation into antibody-secreting plasma cells (24–28), although it is not known whether Tfr cell–derived IL-10 can modulate the GC response. Here, we demonstrate that it does modulate the GC response during acute viral infection. These findings reveal that Tfr cells play a multifaceted role in the fine-tuning of the GC response and are capable of promot-ing or suppressing the GC response depending on the signals provided and the context in which these signals are received.

RESULTSTreg cell–derived IL-10 promotes B cell differentiation and GC developmentTo examine IL-10 production by Tfr cells, we infected 10BiT IL-10 re-porter mice acutely with the Armstrong strain of LCMV and assessed Tfr cell phenotype and kinetics. These reporter mice have a bacterial artificial chromosome transgene containing the Il10 gene locus, with the Thy1.1 complementary DNA (containing a stop codon) replacing the endogenous coding segment of exon 1 of the Il10 locus such that

1Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA. 2Department of Internal Medicine (Rheumatology), Yale University School of Medicine, New Haven, CT 06520, USA. 3Interdepartmental Program in Computational Biology and Bioinformatics, Yale University School of Medicine, New Haven, CT 06520, USA. 4Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA.*These authors contributed equally to this work.†Present address: Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.‡Corresponding author. Email: [email protected]

Copyright © 2017The Authors, somerights reserved;exclusive licenseeAmerican Associationfor the Advancementof Science. No claimto original U.S.Government Works

by guest on Septem

ber 25, 2020http://im

munology.sciencem

ag.org/D

ownloaded from

Page 2: IMMUNE REGULATION Copyright © 2017 Interleukin-10 from CD4 ...€¦ · Laidlaw et al., Sci. Immunol. 2, eaan4767 2017 20 October 2017 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 10

Laidlaw et al., Sci. Immunol. 2, eaan4767 (2017) 20 October 2017

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

2 of 10

cells transcribing its mRNA express Thy1.1 on their cell surface (29). Our previous work validated that Thy1.1+ cells produce IL-10 during LCMV infection (30). Although Tfr cell numbers initially declined af-ter LCMV infection, their number increased from day 5 onward, mir-roring the kinetics of Tfh and pre-Tfh cells and GC B cells [Fig. 1A, gated as in fig. S1, A and B, and as previously described (31–35)]. The ratio of Tfr cells to Tfh cells or GC B cells peaked at day 5 after infection and progressively declined at days 8 and 12 as the increase in Tfh cell and GC B cell numbers outpaced that of Tfr cells (Fig. 1B) (9, 10). There was an increased percentage of Thy1.1+ cells within the Tfr cell population relative to non-Tfr Treg cells at days 5 and 8 after infection, suggesting that IL-10 secretion may be a mechanism by which Tfr cells regulate the emerging GC response within the follicle (Fig. 1C). De-tection of Thy1.1+ Treg cells within the GC by immunofluorescence was impeded because of technical complications related to the dis-rupted splenic architecture at days 5 and 8 after infection and the dim-ness of Thy1.1 expression. Only a small percentage of Tfh cells or GC B cells were competent to express IL-10, with their percentages de-clining over time (fig. S1C). They also made less of this cytokine on a per-cell basis relative to Tfr and non-Tfr Treg cells, suggesting that these cells, in comparison with Tfr or Treg cells, are not an important source of IL-10 for the GC response.

To evaluate the importance of Treg cell–derived IL-10 in the GC response, we generated mice that specifically deleted Il10 in Foxp3- expressing Treg cells, as previously described (29). Il10f/fFoxp3-Cre mice displayed no sign of overt disease at steady state and did not show any consistent trend toward differences in basal percentages of GC B cells, Tfh cells, or Tfr cells (fig. S2). There was also no difference in the Tfr-to-Tfh and Tfr-to-GC B cell ratios in the spleen, peripheral and mesenteric lymph nodes, and Peyer’s patches. We evaluated the B cell response after acute LCMV infection of these animals in comparison with IL-10 intact ones. Although this response appeared similar in the two groups at day 8 after infection, a statistically significant decrease in the per centage and number of B cells with an activated phenotype (IgDlo) and of those cells with a GC phenotype (GL7+CD95+) emerged

over time in mice in which Treg cells lacked Il10 expression, with this dif-ference peaking at day 15 after infection (Fig. 2A and fig. S3A). These mutant mice also displayed reduced GC size as determined by confocal imaging of splenic sections (Fig. 2B). We found that mice lacking Treg cell–derived IL-10 had a reduced percentage and number of plasmablasts (CD138+B220int cells) compared with their IL-10 intact counterparts, albeit a similar number of cells with a GC-dependent memory B cell phenotype (B220+IgDloGL7−CD38+CD95+ cells) (Fig. 2C) (36, 37). They also had reduced serum titers of LCMV-specific IgG2a and IgG1 (Fig. 2D), with a similar number of LCMV-specific memory B cells (Fig. 2E). No defects in GC B cell or plasmablast numbers were evident in mice immunized with NP-OVA [(4-hydroxy-3-nitrophenyl)acetyl- ovalbumin] in complete Freund’s adjuvant (fig. S3B). There were also no apparent defects in GC B cell or plasmablast numbers in LCMV- infected IL-10−/− mice or in mice treated with an anti–IL-10 antibody, suggesting that IL-10 production by different cell types may play op-posing roles in the regulation of the GC response (fig. S4, A and B) (38). Together, these data indicate that IL-10 specifically produced by Treg cells is important in promoting plasmablast differentiation and the development of the GC after viral infection.

Tfr cell–derived IL-10 promotes the GC responseThese findings did not distinguish between the roles of Tfr cell–derived IL-10 and IL-10 produced by Bcl6− Treg cells in regulating the GC re-sponse. Follicular nonresident Treg cells can modulate the GC response as early as day 3 after immunization and likely serve as precursors for Tfr cells (14). To discriminate between these possibilities, mixed bone marrow chimeras (mBMCs) were generated, in which Il10 was specifically ablated in Bcl6-expressing Tfr cells via generation of 50:50 Il10f/fFoxp3-Cre:Bcl6f/fFoxp3-Cre and control 50:50 Il10f/f:Bcl6f/fFoxp3-Cre chimeras (Fig. 3A) (12). In the absence of Tfr cell–derived IL-10, there was a significantly reduced percentage of IgDlo B cells at day 15 after LCMV infection and reduced percentages of GC B cells and plasmablasts (CD138+B220int) in comparison with control chimeras. The importance of Tfr cell–derived IL-10 was further tested using

0

10

20

30

0.00

0.02

0.04

0.06

0.080.200.250.300.35

0.0

0.1

0.2

0.3

0

1,000,000

2,000,000

3,000,000

4,000,000

0

5000

10,000

15,000

0

500,000

1,000,000

1,500,000

0

200,000

400,000

600,000

800,000

1,000,000

0

500,000

1,000,000

1,500,000

2,000,000

2,500,000

Foxp

3__

____

___

Thy1.1 ________ Thy1.1

____

____

_

Thy1

.1

(%)

C Non-Tfr Treg cellsTfr cells * **

Foxp

3

________

13.3 Tfr cellsNon-Tfr Treg cells

Day0 4 8 12

+

DayDay DayDay

Tfr

cells

(#)

Non

-Tfr

T reg

s (#

)

pre-

T fh

cells

(#)

Tfh

cel

ls (#

)

A

GC

B c

ells

(#)

0 5 8 12 0 5 8 12 0 5 8 12 0 5 8 12Day

0 5 8 12

DayDay

T fr/T

fh c

ell r

atio

T fr/G

C B

cel

l rat

io

0 5 8 12 0 5 8 12

B

23

___ *** ___ ___ ** *** ___ ** ___ ___ ** ***

Fig. 1. Tfr cells robustly secrete IL-10 after acute viral infection. Analysis of the Treg cell response after LCMV infection in IL-10 reporter (10BiT Thy1.1) mice. (A) Quan-tification of the number of Tfr cells, non-Tfr Treg cells, pre-Tfh cells, Tfh cells, and GC B cells at days 0, 5, 8, and 12 after infection. Populations are defined as follows: Tfr cells, CD4+Ly6C−PSGL1loCXCR5hiPD1hiFoxp3+; non-Tfr Treg cells, CD4+CXCR5int-loPD1int-loFoxp3+; pre-Tfh cells, CD4+CD44hiLy6C−PSGL1loCXCR5intPD1intFoxp3−; Tfh cells, CD4+CD44hiLy6C−PSGL1loCXCR5hiPD1hiFoxp3−; and GC B cells, B220+IgDloGL7+CD95+. (B) Quantification of the ratio of Tfr cells to Tfh cells or GC B cells at days 0, 5, 8, and 12 after infection. (C) Representative plot of IL-10 expression (assessed as Thy1.1) by Tfr cells (left) and non-Tfr Treg cells (middle) from mice as described in (A). Right: Frequency of Thy1.1+ cells in Tfr cells and non-Tfr Treg cells at days 0, 5, 8, and 12 after LCMV Armstrong infection. Statistical analyses were performed using unpaired two-tailed Student’s t test (*P < 0.05; **P < 0.01; ***P < 0.001). Data are from two experiments representative of four experiments with three to six mice per time point after LCMV Armstrong infection.

by guest on Septem

ber 25, 2020http://im

munology.sciencem

ag.org/D

ownloaded from

Page 3: IMMUNE REGULATION Copyright © 2017 Interleukin-10 from CD4 ...€¦ · Laidlaw et al., Sci. Immunol. 2, eaan4767 2017 20 October 2017 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 10

Laidlaw et al., Sci. Immunol. 2, eaan4767 (2017) 20 October 2017

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

3 of 10

0.0

0.5

1.0

1.5

2.0

0

500,000

1,000,000

1,500,000

0.0

0.5

1.0

0.0

0.2

0.4

0.6

0.8

0

5

10

15

0

100,000

200,000

300,000

0

25,000

5,0000

75,000

100,000

125,000

0

5

10

15

20

25

0

20

40

60

80

0

200,000

400,000

600,000

800,000

1,000,000

A

0 102 103 104 105

0

102

103

104

105

0 102 103 104 105

0

103

104

105

0 102 103 104 105

0

102

103

104

105

0 102 103 104 105

0

103

104

105

IgD

____

__

B220 ________C

D95

____

__

GL7 ________

IgD

____

__

B220 ________

CD

95__

____

GL7 ________

20.5

74.1

13.5

40.9

Il10 f/fIl10 Foxp3-Cre f/f

Il10f/f

Il10 f/f

Foxp3-Cre

Il10f/f

Il10 f/f

Foxp3-Cre

Il10f/f

Il10 f/f

Foxp3-Cre

Il10f/f

Il10 f/f

Foxp3-Cre

________ *________ * ________ * ________ *

IgD

o

f B22

0 (

%)

lo+

GC

B c

ells

of

B22

0 I

gD

(%)

lo+

B22

0 I

gD

(#)

lo+

GC

B c

ells

(#)

0

5

10

15

20

Il10f/f

Il10 f/f

Foxp3-Cre

GC

siz

e (m

m )2

________ ***

Il10f/f

Il10 f/f

Foxp3-Cre

Il10f/f

Il10 f/f

Foxp3-Cre

Il10f/f

Il10 f/f

Foxp3-Cre

Il10f/f

Il10 f/f

Foxp3-Cre

________ * ________ **

CD

138

B22

0

(%)

int

+

CD

38 C

D95

GL7

+

+

of B

220

IgD

(%

)lo

+

-

CD

138

B22

0

(#)

int

+

CD

38 C

D95

GL7

(#)

+

+-

________ *

Il10f/f

Il10 f/f

Foxp3-Cre

Il10f/f

Il10 f/f

Foxp3-Cre

LCM

V-sp

ecifi

c Ig

G2a

(a.u

.)

LCM

V-s

peci

fic Ig

G1

(a.u

.)

B

C

D

Il10 f/fIl10 Foxp3-Cre f/f

________ n.s.

Il10f/f

Il10 f/f

Foxp3-Cre

Il10f/f

Il10 f/f

Foxp3-Cre

IgG

2a m

emor

y B

cel

ls (#

)

0

500

1000

1500

LCM

V-s

peci

fic

0

500

1000

1500

IgG

1 m

emor

y B

cel

ls (#

) LC

MV

-spe

cific

E

IgD PNA

Fig. 2. Regulatory CD4+ T cell–derived IL-10 is important for B cell differentiation and the GC response. Analysis of the B cell response in Il10f/f and Il10f/fFoxp3-Cre mice 15 days after LCMV infection. (A) Top: Representative plots of the B cell responses in Il10f/f or Il10f/fFoxp3-Cre mice. Bottom: Frequency and number of cells as indi-cated by the gates shown above. (B) Representative confocal images of the GC from Il10f/f or Il10f/fFoxp3-Cre mice. Right: GC sizes as measured by ImageJ. The sections were taken from four mice of each genotype. Scale bar, 100 m. (C) Left: Frequency of plasmablasts and memory B cells defined by the expression of surface markers. Right: Absolute numbers of plasma cells and memory B cells. (D) Enzyme-linked immunosorbent assay quantification of LCMV-specific IgG2a and IgG1 antibody levels, indicated by arbitrary units (a.u.). All the analyses of the GC response were performed 15 days after acute LCMV Armstrong infection. n.s., not significant. (E) Enzyme-linked immunospot quantification of the number of LCMV-specific IgG2a and IgG1 memory B cells. All the analyses of the GC response were performed 60 days after acute LCMV Armstrong infection. Statistical analyses were performed using unpaired two-tailed Student’s t test (*P < 0.05; **P < 0.01; ***P < 0.001). Data for (A) to (C) are from one experiment representative of three experiments with three to five mice per group carried out 15 days after LCMV Armstrong infection and from four experiments with three to five mice per group carried out 12 days after LCMV Armstrong infection. Data for (D) are pooled from two experiments carried out 12 or 15 days after infection. Data for (E) are pooled from two experiments with five to seven mice per group carried out 60 days after infection.

by guest on Septem

ber 25, 2020http://im

munology.sciencem

ag.org/D

ownloaded from

Page 4: IMMUNE REGULATION Copyright © 2017 Interleukin-10 from CD4 ...€¦ · Laidlaw et al., Sci. Immunol. 2, eaan4767 2017 20 October 2017 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 10

Laidlaw et al., Sci. Immunol. 2, eaan4767 (2017) 20 October 2017

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

4 of 10

another approach in which mBMCs were generated using SAP- deficient (SAP−/−) mice in place of Bcl6f/fFoxp3-Cre (Fig. 3B). SAP is required for Tfr cell differentiation (8, 9). Again, we found that loss of Tfr cell–derived IL-10 resulted in decreased percentages of IgDlo B cells, GC B cells, and plasmablasts. These data indicate that Tfr cell–derived IL-10 supports B cell differentiation and the GC response.

IL-10 signals via B cells to promote the GC responseThere are multiple pathways by which Treg cell–derived IL-10 could modulate the GC response, including acting directly on B cells or indirectly through Tfh cells or dendritic cells (DCs) (39, 40). To assess which cell populations were responsive to IL-10 signaling, we stimu-lated splenocytes from LCMV-infected mice with IL-10 and deter-mined phospho-STAT3 (pSTAT3) in GC B cells, DCs, and Tfh cells. Although both GC B cells and DCs displayed robust pSTAT3 expres-sion relative to unstimulated controls, Tfh cells were poorly responsive, suggesting that IL-10 does not act directly upon them to regulate the GC response (Fig. 4A). Consistent with this finding, we determined that the LCMV-specific CD4+ T cell response against the immuno-dominant GP66–77 epitope was similar be tween control mice and those lacking Treg cell–derived IL-10 at day 8 after infection (fig. S5A). The LCMV-specific T helper 1 (TH1) (CD44hiPSGL1hiLy6C+ cells) and Tfh (CD44hiPSGL1loLy6C−CXCR5+PD1+ cells) responses, as well as the percentages of Treg (Foxp3+CD4+ cells) and Tfr cells, were also similar between the groups (fig. S5B) (31).

To identify the cell population that IL-10 was acting upon to reg-ulate the GC response, we generated mice in which the Il10ra gene was deleted specifically on B cells, DCs, or T cells (22). The B cell re-sponse in these mice was then assessed at day 15 after LCMV infection. Although the B cell response in mice in which T cells could not sense IL-10 was comparable with control mice, Il10rf/fCd19-Cre mice dis-played a significant decrease in the percentages of activated and GC

B cells compared with Il10rf/f controls (Fig. 4B). Il10rf/fCd11c-Cre mice also had a reduced percentage of IgDlo B cells but an equivalent fraction of these cells with a GC phenotype, indicating that although IL-10 may partially act through DCs to promote B cell activation, this pathway does not account for the defect in the GC response (Fig. 4B). Together, these findings suggest that Treg cell–derived IL-10 can act directly on B cells to promote their activation and differentiation into GC B cells.

Treg cell–derived IL-10 drives GC B cells to adopt a dark zone phenotypeTo help understand how Treg cell–derived IL-10 influenced the fate of GC B cells, we performed RNA sequencing (RNA-seq) analysis to evaluate the gene expression profile of GC B cells isolated from Il10f/fFoxp3-Cre and control Il10f/f mice at day 12 after LCMV infection. There were 138 differentially expressed genes (DEGs) in GC B cells between the two groups, with 84 genes up-regulated and 54 genes down- regulated in the Il10f/fFoxp3-Cre mice compared with controls (Padj < 0.1) (Fig. 5A and fig. S6A). Among the DEGs were a number of genes used to distinguish dark zone and light zone GC B cells, including Cxcr4 and Cd83 (6, 41). Gene set enrichment analysis (GSEA) was used to detect genome-wide changes and identified a significant enrich-ment for a light zone GC B cell signature in GC B cells isolated from Il10f/fFoxp3-Cre mice (Fig. 5, B and C, and fig. S6B). The dark zone is the proliferative compartment of the GC and the predominant site of Ig gene somatic hypermutation, in contrast to the light zone where GC B cells have a largely nonproliferative state and compete for the T cell help necessary to induce dark zone reentry (42–44). GC B cells isolated from Il10f/fFoxp3-Cre compared with control mice were also highly impaired in gene sets associated with translational activity (Fig. 5C). Together, these findings suggest that Treg cell–derived IL-10 promotes GC B cells to adopt a dark zone phenotype.

0

10

20

30

40

50

________ *________ ** ________ *

IgD

o

f B22

0 (

%)

lo+

GC

B c

ells

of

B22

0 I

gD

(%)

lo+

0

5

10

15

20

25

0

10

20

30

40

0

1

2

3

4

5C

D13

8 B

220

(%

)in

t+

IgD

o

f B22

0 (

%)

lo+

GC

B c

ells

of

B22

0 I

gD

(%)

lo+

CD

138

B22

0

(%)

int

+

0

5

10

15

0

1

2

3

4________ *________ *** ________ *

Il10f/f

Il10 f/f

Foxp3-CreSAP : -/- Il10

f/fIl10

f/f Foxp3-Cre

Il10f/f

Il10 f/f

Foxp3-Cre

Bcl6 f/f

Foxp3-Cre : Il10

f/fIl10

f/f Foxp3-Cre

Il10f/f

Il10 f/f

Foxp3-Cre

Il10f/f

Il10 f/f

Foxp3-Cre

A

B

Donors

Bcl6f/f

Foxp3-Cre

(Tfr cell−deficient)

Il10f/f

Foxp3-Cre

Il10f/f

IL-10 T cells

Irradiated recipients

8 weeks+/+

fr

IL-10 T cells-/- fr

8 weeks

Donors

(Tfr cell–deficient)

Il10f/f

Foxp3-Cre

Il10f/f

IL-10 T cells

Irradiated recipients

8 weeks+/+

fr

IL-10 T cells-/- fr

8 weeksSAP mice-/-

Fig. 3. Tfr cell–derived IL-10 is important for B cell differentiation and the GC response. (A) Analysis of the B cell response at day 15 after acute LCMV Armstrong in-fection in 50:50 Il10f/f/Bcl6f/fFoxp3-Cre or Il10f/fFoxp3-Cre/Bcl6f/fFoxp3-Cre mBMCs. Left: Schematic for the experiment. Right: Data are pooled from two experiments with four to five mice per group carried out 15 days after LCMV Armstrong infection. (B) Analysis of the B cell response at day 15 after acute LCMV Armstrong infection in 50:50 Il10f/f/SAP−/− or Il10f/fFoxp3-Cre/SAP−/− mBMCs. Left: Schematic for the experiment. Right: Data are from one experiment representative of two experiments with four to nine mice per group carried out 15 days after LCMV Armstrong infection. Statistical analyses were performed using unpaired two-tailed Student’s t test (*P < 0.05; **P < 0.01; ***P < 0.001).

by guest on Septem

ber 25, 2020http://im

munology.sciencem

ag.org/D

ownloaded from

Page 5: IMMUNE REGULATION Copyright © 2017 Interleukin-10 from CD4 ...€¦ · Laidlaw et al., Sci. Immunol. 2, eaan4767 2017 20 October 2017 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 10

Laidlaw et al., Sci. Immunol. 2, eaan4767 (2017) 20 October 2017

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

5 of 10

To more directly test this hypothesis, we determined the dark zone and light zone phenotypes of GC B cells from control mice and those lacking Treg cell–derived IL-10. In agreement with our RNA-seq data, there was a small but significant decrease in the percentage of dark zone GC B cells and an increase in that of light zone GC B cells in GC B cells from Il10f/fFoxp3-Cre mice (Fig. 5D). Aligning with previous results, we did not detect a difference in GC B cell pro-liferation or survival between the groups (fig. S7) (43, 44). These data support a model in which Treg cell–derived IL-10 acts on light zone GC B cells to skew them toward a dark zone phenotype. These data

do not discount the possibility that Treg cell–derived IL-10 acts on B cells during the early stages of the GC response to influence their ability to later adopt a dark zone phenotype.

Treg cell–derived IL-10 promotes dark zone phenotype through induction of nuclear FOXO1We next analyzed our RNA-seq data using Integrated Pathway Anal-ysis (IPA; Ingenuity Systems, www.ingenuity.com). This analysis re-vealed that eukaryotic initiation factor 2 (eIF2) signaling was the most significantly dysregulated pathway in Il10f/fFoxp3-Cre mice compared

0

20

40

60

___

No stimIL-10

pSTAT3 _________________________________________________________

Max

(%)

GC B cells Dendritic cells Tfh cells

IgD

____

__

B220 ________

CD

95__

____

GL7 ________Ig

D__

____

B220 ________

CD

95__

____

GL7 ________

Il10rα f/fIl10rα Cd19-Cre f/f

0 102 103 104 105

0

102

103

104

105

<Pacific Blue-A>: IgD

0 102 103 104 105

0

102

103

104

105

<PE-Cy7-A>: CD95

0 102 103 104 105

0

102

103

104

105

<Pacific Blue-A>: IgD

0 10 2 10 3 10 4 10 5

0

10 3

10 4

10 5

<PE-Cy7-A>: CD95

26.5

42.9

18.7

28.3

IgD

____

__

B220 ________

CD

95__

____

GL7 ________

IgD

____

__

B220 ________

CD

95__

____

GL7 ________

Il10rα Cd11c-Cre f/fIl10rα Cd4-Cre f/f

0 102 103 104 105

0

102

103

104

105

<Pacific Blue-A>: IgD

0 102 103 104 105

0

102

103

104

105

<PE-Cy7-A>: CD95

0 102 103 104 105

0

102

103

104

105

<Pacific Blue-A>: IgD

0 102 103 104 105

0

102

103

104

105

<PE-Cy7-A>: CD95

18.2

42.8

27.8

42.3

0

10

20

30

IgD

o

f B22

0 (

%)

lo+

Il10rαf/fIl10rα

f/f Cd19-Cre

Il10rα f/f

Cd11c-Cre

Il10rα f/f

Cd4-Cre

Il10rα Il10rα

Cd19-Cre

Il10rα

Cd11c-Cre

Il10rα

Cd4-Cre

GC

B c

ells

of

B22

0 I

gD

(%)

lo+

________ *________________ ** ________ *

A

B

0 102 103 104 1050

20

40

60

80

100

0 102 103 104 1050

20

40

60

80

100

0 102 103 104 1050

20

40

60

80

1002301 72937 11

2700 251664 10

657 20466 26

+-+-

+-+-

+-+-

Fig. 4. IL-10 acts on B cells to regulate the GC response. (A) GC B cells and DCs, but not Tfh cells, are responsive to IL-10. Analysis of IL-10 responsiveness in GC B cells, DCs (CD11chiMHCII+), and Tfh cells. Splenocytes isolated from mice at day 12 after LCMV infection were stimulated for 30 min with IL-10, and pSTAT3 levels were determined. Data are from one experiment representative of three experiments with four mice per group. (B) Analysis of the B cell response in Il10rf/f, Il10rf/fCd19-Cre, Il10rf/fCd11c-Cre, and Il10rf/fCd4-Cre mice 15 days after LCMV infection. Top: Representative plots of B cell response. Bottom: Quantification of B cell response shown above. Statistical analyses were performed using unpaired two-tailed Student’s t test (*P < 0.05; **P < 0.01). Data are from one experiment representative of two to three experiments with three to seven mice per group carried out 15 days after LCMV Armstrong infection. Mice for the control Il10rf/f group were pooled from littermate controls produced from the inde-pendent crosses used to generate the experimental groups.

by guest on Septem

ber 25, 2020http://im

munology.sciencem

ag.org/D

ownloaded from

Page 6: IMMUNE REGULATION Copyright © 2017 Interleukin-10 from CD4 ...€¦ · Laidlaw et al., Sci. Immunol. 2, eaan4767 2017 20 October 2017 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 10

Laidlaw et al., Sci. Immunol. 2, eaan4767 (2017) 20 October 2017

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

6 of 10

with controls (Fig. 6A), in agreement with our GSEA indicating im-paired translation. mTOR signaling was also significantly up-regulated (Fig. 6A). Analysis of potential upstream regulators identified im-paired Forkhead box protein 1 (FOXO1) and enhanced RICTOR (rapamycin-insensitive companion of mTOR) signaling, an mTOR signaling component, as the most significant regulators of the gene signature (Fig. 6B). The transcription factor FOXO1 and PI3K (phos-phatidylinositol 3-kinase) signaling, an inducer of mTOR, execute opposing roles in the regulation of the dark zone and light zone GC B cell fate, with ablation of FOXO1 or induction of PI3K activity result-ing in a loss of the dark zone phenotype cells (42, 43, 45). Thus, regu-lation of FOXO1 activity by Treg cell–derived IL-10 could modulate

the dark zone phenotype of GC B cells. To test this hypothesis, we determined total FOXO1 expression at day 15 after LCMV infection in GC B cells from Il10f/fFoxp3-Cre mice and identified decreased protein levels relative to controls (Fig. 6C). This decrease was evident in both dark zone and light zone GC B cells (fig. S8A). We also found that GC B cells from Il10f/fFoxp3-Cre mice displayed increased pS6 and pAktS473 expression (fig. S8B). Collectively, these data indicated that Treg cell–derived IL-10 is necessary for maximal FOXO1 induc-tion in GC B cells.

We next asked whether IL-10 promotes FOXO1 nuclear translo-cation in activated B cells. Active FOXO1 localizes to the nucleus where it can instruct a specific gene program (46). To mirror the in vivo

0

20

40

60

0

10

20

30

40

0

500

1000

1500

A

−0.6

−0.4

−0.2

0

0.2

0.4

0.6

log2( )

Il10 f/f

0 5000 10,000 15,000 20,000

Rank in ordered dataset

Enr

ichm

ent s

core

(ES

)

Light zone (G)Dark zone (G)

−2 −1 2

FWER

< 1e−05

1e−04

0.001

0.01

0.1

Light zone (G)

Dark zone (G)

Metabolismof RNA (R)

Metabolismof mRNA (R)

Translation (R)

Respiratoryelectron

transport (R)SRP-dependent

proteinTargeting (R)

Formation ofthe ternary

complex (R)

Nonsensemediated decay (R)

3’UTR mediatedtranslational

regulation (R)

Peptide chainelongation (R)

Ribosome (K)

Normalized enrichment score

0 1

Il10 Foxp3-Cref/f

Il10f/f

Il10 Foxp3-Cref/f

B

Il10 Foxp3-Cref/f Il10 f/f

C

Il10 f/fIl10 Foxp3-Cref/f

Rplp0

Cnot6

Psip1

Rpl37

Cxcr4

Malat1

Ccnl1

Eif3f

Rock2

Suz12

Mki67

Pgls

Chchd10

Apoe

Nfkbia

Icam1

Dusp1

Tapbp

Il21r

Stat6

Cd83

Ccr6

Grn

Ltb

Il4i1

D

CX

CR

4__

___

CD86 ________________________

Il10 f/f

0 102 103 104 105

0

102

103

104

105

Il10 Foxp3-Cre f/f

0 102 103 104 105

0

102

103

104

105

+C

XC

R4

CD

86 (

%)

-

-C

XC

R4

CD

86 (

%)

+

CX

CR

4 M

FI

Il10f/f

Il10 f/f

Foxp3-Cre

Il10f/f

Il10 f/f

Foxp3-Cre

Il10f/f

Il10 f/f

Foxp3-Cre

_______ * _______ * _______ *

53.7

34

41.3

37

0.6

0

0.3

–0.3

Fig. 5. GC B cells in mice lacking regulatory CD4+ T cell–derived IL-10 display an enhanced light zone GC B cell gene signature. (A) RNA-seq analysis of select DEGs among mRNA isolated from GC B cells pooled from Il10f/f and Il10f/fFoxp3-Cre mice 12 days after LCMV Armstrong infection presented as expression (log2) in Il10f/fFoxp3-Cre cells relative to that in Il10f/f cells (key below; columns indicate paired replicates). (B) GSEA of light zone and dark zone signatures in GC B cells, based on published gene sets (6, 41). A positive enrichment score (ES) signifies enrichment in the Il10f/fFoxp3-Cre sample relative to the Il10f/f condition of a given gene set, that is, more highly ex-pressed. (C) Normalized ES (NES) for select pathways identified from the Reactome Pathway Database (R), Kegg Pathway Database (K), and published gene sets (G), where dot size represents P values adjusted by the family-wise error rate (FWER). All gene sets attain significant enrichment [false discovery rate (FDR) < 0.001], with the excep-tion of the dark zone gene set (FDR = 0.5). Data are from three independent experiments with three mice per group pooled for each sample. (D) Analysis of the light zone and dark zone GC B cell response in Il10f/f and Il10f/fFoxp3-Cre mice 15 days after LCMV infection. Representative plots (left) of GC B cells as gated in fig. S1B. The numbers of the outlined area indicate dark zone (top left) and light zone (bottom right) based on the expression of the surface markers CXCR4 and CD86. Middle: Frequency of GC B cells from light zone gate defined by CXCR4+CD86− and dark zone gate defined by CXCR4−CD86+ from Il10f/f or Il10f/fFoxp3-Cre mice. Right: Expression of CXCR4 in GC B cells. Statistical analyses were performed using unpaired two-tailed Student’s t test (*P < 0.05). Data are from one experiment representative of two experiments with at least four mice per group carried out 15 days after LCMV Armstrong infection. MFI, mean fluorescence intensity.

by guest on Septem

ber 25, 2020http://im

munology.sciencem

ag.org/D

ownloaded from

Page 7: IMMUNE REGULATION Copyright © 2017 Interleukin-10 from CD4 ...€¦ · Laidlaw et al., Sci. Immunol. 2, eaan4767 2017 20 October 2017 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 10

Laidlaw et al., Sci. Immunol. 2, eaan4767 (2017) 20 October 2017

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

7 of 10

context in which B cells are exposed to IL-10, cells were isolated from mice at day 5 after LCMV infection and cultured with or with-out IL-10. Day 5 coincides with the peak expression of IL-10 by Tfr cells, a major source of IL-10 within the inner follicle (Fig. 1C and fig. S1C), and allowed us to assess FOXO1 nuclear translocation in cells that have not yet had prolonged exposure to IL-10. Using Amnis ImageStream analysis, we observed an increase in the percentage of nuclear translocated FOXO1 in activated B cells (Fig. 6D). IL-10 did not induce nuclear translocation of FOXO1 in follicular B cells, de-spite robust pSTAT3 signaling (fig. S8C), suggesting that IL-10 acts in concert with other signals received by activated B cells, such as B cell receptor (BCR), costimulation, and cytokines, to promote FOXO1 activity.

In addition to regulating dark zone reentry, FOXO1 is also im-portant in promoting Ig affinity maturation (42, 43). To assess af-finity maturation, GC B cells from control Il10f/f and experimental Il10f/fFoxp3-Cre mice were sorted at day 15 after LCMV infection and a large number of the Ig heavy chain variable region (VH) genes were cloned and sequenced using a pooled primer approach, as previously described (47). The overall mutation frequency in the complementarity- determining region (CDR) was similar between groups (Fig. 6E). However, we identified a reduction in the degree of positive selection within the CDR in the Il10f/fFoxp3-Cre mice, which was absent in the framework region (FWR) (Fig. 6E). The trend was observed in two

independent experiments but did not reach statistical significance be-cause of the limitation of the number of clones obtained. We further examined the amino acid physicochemical properties of CDR3 be-cause diversity in this region is a key determinant of antigen binding. Mice that lacked Treg cell–derived IL-10 displayed a trend toward a higher grand average of hydrophobicity (GRAVY) score, mean side-chain bulkiness, and abundance of aromatic residues in this region, consistent with the notion that less selection is occurring in these cells because positively selected cells typically display reduction in these metrics (fig. S9) (48).

DISCUSSIONUnderstanding the signals regulating GC B cell differentiation is critical for the development of targeted therapies that can modulate GC output. Here, we describe a role for Tfr cells in promoting the GC response through local production of IL-10 during viral infection. IL-10–secreting Treg cells act on B cells to drive their differentiation into plasmablasts and GC B cells. Both IL-10 expression and the Tfr-to-GC B cell ratio peak at day 5 after infection, suggesting that Tfr cell–derived IL-10 may be predominantly acting early to drive B cell differentiation. Tfr cell–derived IL-10 promotes expression and activ-ity of FOXO1, thereby facilitating adoption of a dark zone pheno-type by GC B cells and potentially enhancing affinity maturation.

A IPA canonical pathways

–4–2

4 P value

<1e−05

<0.01

<0.05

FOXO1 MYC CD40 EGFR Jnk Ifnar CD40LG SYVN1 TNF Cg IFNG RICTOR

Activation z score

02

Il10f/f

Il10 Foxp3-Cref/f

B IPA upstream regulators

Regulation of eIF4 and p70S6K signalingeIF2 signaling

mTOR signaling

Dendritic cell maturation

Protein ubiquitination pathway

– log (B-H P value)

Threshold

40 2 106 8 12

E

Il10f/f

Il10 f/f

Foxp3-Cre

Mut

atio

ns in

CD

R (#

)

Selection strength (Σ)

Den

sity

___ Il10 f/fIl10 Foxp3-Cre f/f

____

____

__

–1.0 –0.5 0.0 0.5 1.0

0.0

2.5

5.0

7.5

2.5

5.0

7.5

CD

RFW

R

0

5

10

15

0

1000

2000

3000

4000

5000

Il10f/f

Il10 f/f

Foxp3-Cre

FOX

O1

MFI

_______ **

C

0 102 103 104 1050

20

40

60

80

100

0

2

4

6

D

No stim. IL-10–stim.

Nuc

lear

tran

sloc

ated

FOX

O1

of Ig

D

(%)

*

lo

_______

GC B cells

FOXO1 ___________

Max

(%)

____

____

__

Fig. 6. Reduced FOXO1 expression in GC B cells from mice lacking regulatory CD4+ T cell–derived IL-10. (A) IPA canonical pathway analysis. The number on the x axis indicates the negative log P values adjusted by the Benjamini-Hochberg (B-H) procedure, which is the significance of the probability that the genes in the data set de-scribed here are associated with the canonical pathway listed. The orange line represents the threshold of significance for a B-H multiple testing correction P value of 0.05. (B) IPA upstream regulator analysis. The top upstream regulators that are predicted to be responsible for the gene expression changes observed in the Il10f/fFoxp3-Cre sample relative to the Il10f/f sample are shown. A negative activation z score indicates that the upstream regulator is predicted to be inhibited in the Il10f/fFoxp3-Cre sample relative to the Il10f/f sample, and a positive score indicates that the upstream regulator is predicted to be activated. (C) Expression of FOXO1 in GC B cells from Il10f/f or Il10f/fFoxp3-Cre mice 15 days after LCMV Armstrong infection. Data are from one experiment representative of two experiments with at least four mice per group carried out 15 days after LCMV Armstrong infection. (D) Assessment of the percentage of nuclear translocated FOXO1 after IL-10 stimulation in IgDlo B cells 5 days after LCMV infection as determined by Amnis ImageStream. Statistical analyses were performed using paired two-tailed Student’s t test (*P < 0.05; **P < 0.01). Data are pooled from three experiments with four mice per group carried out 5 days after LCMV Armstrong infection. (E) Assessment of the number of mutations in the CDR region (left) and selection strength (right) in both the CDR region and FWR of a large number of the VH genes of GC B cells from Il10f/f or Il10f/fFoxp3-Cre mice 15 days after LCMV infection. Data are pooled from two experiments with three to four mice per group carried out 15 days after LCMV Armstrong infection.

by guest on Septem

ber 25, 2020http://im

munology.sciencem

ag.org/D

ownloaded from

Page 8: IMMUNE REGULATION Copyright © 2017 Interleukin-10 from CD4 ...€¦ · Laidlaw et al., Sci. Immunol. 2, eaan4767 2017 20 October 2017 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 10

Laidlaw et al., Sci. Immunol. 2, eaan4767 (2017) 20 October 2017

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

8 of 10

These results illustrate the complex role of Tfr cells in the regulation of the GC response and the need to consider both the suppressive and stimulatory roles of Treg cells in control of humoral immunity.

How do Tfr cells balance their suppression and promotion of GC output? IL-10–competent Tfr cells express high levels of CTLA4, so it is unlikely that IL-10–secreting and CTLA4-expressing Tfr cells repre-sent unique populations. One possible explanation is that suppression by Tfr cells is reliant on cell contact (e.g., CTLA4-CD80/86 interactions). In such a scenario, light zone GC B cells engaged in productive major histocompatibility complex (MHC)–peptide–T cell receptor (TCR) ex-change with Tfh cells would be less available for this type of interaction, thus allowing IL-10 secretion by nearby Tfr cells to stimulate GC B cell differentiation and dark zone reentry. However, GC B cells not engaged with Tfh cells would be available to interact with Tfr cells, where the suppressive function of CTLA4 could dominate the stimulatory role of IL-10. In this manner, Tfr cells could promote the continued differ-entiation of affinity-matured, antigen-bearing GC B cells while sup-pressing those cells with low antigen affinity. Alternatively, B cells engaged in productive interactions with Tfh cells, including before GC formation, might be rendered more sensitive to IL-10 signaling (or less susceptible to inhibition by CTLA4), providing signals that initiate dark zone polarization of these cells or their growth advantage. The finding that Tfr cells restrict the outgrowth of non–antigen-specific GC B cells supports these models and suggests an important role for Tfr cells in restraining the expansion of autoreactive GC B cells (9). It will be important for future work to more directly test these ideas and explore other mechanisms by which Tfr cells may modulate the GC response.

Distinguishing between the function of Tfr cells and follicular non-resident Treg cells in modulating the GC response is critical. Ablation of CTLA4 expression on Treg cells impairs CD86 expression on B cells before the formation of Tfr cells, indicating that follicular nonresident Treg cells influence the GC response (14). It is likely that there are multiple mechanisms by which Treg cells could exert this effect, in-cluding regulation of T or B cell avidity and the activation state of DCs (22, 23, 49). Here, we found that depletion of Tfr cell–derived IL-10 led to an impaired GC response. This finding does not rule out a contribution of IL-10–producing follicular nonresident Treg cells on the regulation of the GC response. It also does not distinguish between the roles of Tfr cells inside and outside of the GC, as well as those cells present within the follicle before GC development.

Tfr cell–derived IL-10 likely contributes to the GC B cell response through multiple mechanisms. Our work reveals its promotion of FOXO1 expression, which is required for dark zone formation be-cause of its essential role in gene program instruction, including the up-regulation of the chemokine receptor CXCR4 required for GC B cell migration into the dark zone (42–45). However, although regu-lation of FOXO1 activity may explain the dysregulated dark zone phenotype and affinity maturation of GC B cells from mice lacking Treg cell–derived IL-10, it does not explain the decrease in GC B cell number and output. GC B cell proliferation and somatic hypermutation typically occur in the dark zone, but these processes are maintained even in cells that are unable to access the dark zone (42–44). Therefore, it is likely that Tfr cell–derived IL-10 functions in a FOXO1-independent manner to regulate B cell differentiation and GC development.

The IL-10–STAT3 pathway has been implicated as a promoter of diffuse large B cell lymphoma (DLBCL) and systemic lupus erythema-tosus (50–53). A high level of circulating IL-10 and active intracel-lular STAT3 are associated with clinically aggressive cases of DLBCL

(51, 52). Constitutively active STAT3 promotes cell proliferation and survival, and anti–IL-10R antibody treatment induces cell cycle arrest and apoptosis in DLBCL cell lines (50, 52). Among known STAT3 tar-gets that are inhibited by IL-10R blockade are genes involved in cell cycle progress, antiapoptotic factors, and proto-oncogenes, including Ccnd1, Mcl1, Junb, and cMyc (50, 52). We found a robust impairment in pathways associated with RNA translation in mice lacking Treg cell– derived IL-10, suggesting that IL-10–driven STAT3 may regulate GC B cell proliferation and differentiation by inducing the transla-tion of mRNAs encoding proliferation-promoting proteins or pro-teins involved in differentiation (54). It will be important to elucidate which genes IL-10–driven STAT3 is acting upon to drive B cell prolif-eration and differentiation during viral infection and the extent that this pathway overlaps with disease progression in individuals with DLBCL. Therapies designed to modulate the stimulatory potential of Tfr cells could prove an effective method to bolster humoral immu-nity or restrain the expansion of autoreactive or malignant B cell clones.

MATERIALS AND METHODSStudy designThe aim of this study was to characterize the role of Tfr cell–derived IL-10 in the regulation of the GC response after acute viral infection. Most of the experiments consisted of enumerating population frequen-cies by flow cytometry in different genetic mouse models, analysis of GCs by immunofluorescence, analysis of RNA-seq data, and quantifi-cation of nuclear localized transcription factors. Littermate comparisons were used for all experiments where possible. Control and experimental groups were age- and sex-matched. The investigators were not blinded. Experimental replications are indicated in the figure legends.

MiceC57BL/6 mice were purchased from the National Cancer Institute or the Jackson Laboratory. B6.129P2-Il10tm1Cgn/J (Il10−/−), B6.129(cg)- Foxp3tm3(DTR/GFP)Ayr/J (Foxp3GFP-DTR), B6.129S(FVB)-Bcl6tm1.1Dent/J (Bcl6f/f), B6.129S6-Sh2d1atm1Pls/J (SAP−/−), and B6.129(Cg)- Foxp3tm4(YFP/cre)Ayr/J (Foxp3-Cre) mice were purchased from the Jackson Laboratory. 10BiT mice (29), Il10f/f mice (55), and Cd4-Cre, Cd19-Cre, and Cd11c-Cre mice have been described. Il10rf/f mice were generated by the Flavell laboratory, as previously described (22). All animal experiments were done with approval of the Yale Institutional Animal Care and Use Committee.

Infection and treatmentsMice were given intraperitoneal administration of 2 × 105 plaque- forming units of LCMV Armstrong. Diphtheria toxin was reconstituted according to the manufacturer’s instructions (Sigma). Mice were given diphtheria toxin at a dose of 50 g per kilogram of body weight on days 4 and 5 (two doses) after infection, as described (56). For IL-10 block-ade, an anti–IL-10 monoclonal antibody (0.25 mg/ml, JES5-2A5 clone, provided by J. M. M. den Haan, VU University Medical Center, Amsterdam, Netherlands) was administered intraperitoneally every other day. For NP-OVA in complete Freund’s adjuvant immunization, mice were given 100 g of NP-OVA mixed with an equal volume of complete Freund’s adjuvant.

Statistical analysisResults represent means ± SEM, unless indicated otherwise. Statistical significance was determined by paired or unpaired Student’s t test.

by guest on Septem

ber 25, 2020http://im

munology.sciencem

ag.org/D

ownloaded from

Page 9: IMMUNE REGULATION Copyright © 2017 Interleukin-10 from CD4 ...€¦ · Laidlaw et al., Sci. Immunol. 2, eaan4767 2017 20 October 2017 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 10

Laidlaw et al., Sci. Immunol. 2, eaan4767 (2017) 20 October 2017

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

9 of 10

Statistical analyses were performed using Prism GraphPad software version 6.0 (*P < 0.05; **P < 0.01; ***P < 0.001).

SUPPLEMENTARY MATERIALSimmunology.sciencemag.org/cgi/content/full/2/16/eaan4767/DC1Supplementary MethodsFig. S1. Flow cytometric gating strategy for Tfh, pre-Tfh, GC B, Treg, and Tfr cells, with IL-10 expression by these populations.Fig. S2. Mice lacking regulatory CD4+ T cell–derived IL-10 do not have defects in steady-state lymphoid cell populations.Fig. S3. Temporal development of the GC B cell response in mice lacking regulatory CD4+ T cell–derived IL-10.Fig. S4. Systemic IL-10 is not required for the GC response.Fig. S5. Regulatory CD4+ T cell–derived IL-10 is not required for effector CD4+ T cell differentiation.Fig. S6. Heat map of DEGs based on RNA-seq.Fig. S7. GC B cells in mice lacking regulatory CD4+ T cell–derived IL-10 display similar levels of proliferation and death.Fig. S8. IL-10 does not induce FOXO1 nuclear translocation in IgDhi B cells.Fig. S9. The VH CDR3 region of GC B cells in mice lacking regulatory CD4+ T cell–derived IL-10 displays altered amino acid physiochemical properties.Table S1. Tabulated data for Figs. 1 to 6 and figs. S1 to S9.References (57–68)

REFERENCES AND NOTES 1. N. S. De Silva, U. Klein, Dynamics of B cells in germinal centres. Nat. Rev. Immunol. 15,

137–148 (2015). 2. K. Basso, R. Dalla-Favera, Germinal centres and B cell lymphomagenesis. Nat. Rev.

Immunol. 15, 172–184 (2015). 3. R. J. Johnston, A. C. Poholek, D. DiToro, I. Yusuf, D. Eto, B. Barnett, A. L. Dent, J. Craft,

S. Crotty, Bcl6 and Blimp-1 are reciprocal and antagonistic regulators of T follicular helper cell differentiation. Science 325, 1006–1010 (2009).

4. X. Liu, X. Chen, B. Zhong, A. Wang, X. Wang, F. Chu, R. I. Nurieva, X. Yan, P. Chen, L. G. van der Flier, H. Nakatsukasa, S. S. Neelapu, W. Chen, H. Clevers, Q. Tian, H. Qi, L. Wei, C. Dong, Transcription factor Achaete-Scute homologue 2 initiates follicular T-helper-cell development. Nature 507, 513–518 (2014).

5. S. G. Tangye, C. S. Ma, R. Brink, E. K. Deenick, The good, the bad and the ugly—TFH cells in human health and disease. Nat. Rev. Immunol. 13, 412–426 (2013).

6. G. D. Victora, T. A. Schwickert, D. R. Fooksman, A. O. Kamphorst, M. Meyer-Hermann, M. L. Dustin, M. C. Nussenzweig, Germinal center dynamics revealed by multiphoton microscopy with a photoactivatable fluorescent reporter. Cell 143, 592–605 (2010).

7. A. D. Gitlin, Z. Shulman, M. C. Nussenzweig, Clonal selection in the germinal centre by regulated proliferation and hypermutation. Nature 509, 637–640 (2014).

8. Y. Chung, S. Tanaka, F. Chu, R. I. Nurieva, G. J. Martinez, S. Rawal, Y.-H. Wang, H. Lim, J. M. Reynolds, X.-h. Zhou, H.-m. Fan, Z.-m. Liu, S. S. Neelapu, C. Dong, Follicular regulatory T cells expressing Foxp3 and Bcl-6 suppress germinal center reactions. Nat. Med. 17, 983–988 (2011).

9. M. A. Linterman, W. Pierson, S. K. Lee, A. Kallies, S. Kawamoto, T. F. Rayner, M. Srivastava, D. P. Divekar, L. Beaton, J. J. Hogan, S. Fagarasan, A. Liston, K. G. C. Smith, C. G. Vinuesa, Foxp3+ follicular regulatory T cells control the germinal center response. Nat. Med. 17, 975–982 (2011).

10. I. Wollenberg, A. Agua-Doce, A. Hernández, C. Almeida, V. G. Oliveira, J. Faro, L. Graca, Regulation of the germinal center reaction by Foxp3+ follicular regulatory T cells. J. Immunol. 187, 4553–4560 (2011).

11. M. Aloulou, E. J. Carr, M. Gador, A. Bignon, R. S. Liblau, N. Fazilleau, M. A. Linterman, Follicular regulatory T cells can be specific for the immunizing antigen and derive from naive T cells. Nat. Commun. 7, 10579 (2016).

12. H. Wu, Y. Chen, H. Liu, L.-L. Xu, P. Teuscher, S. Wang, S. Lu, A. L. Dent, Follicular regulatory T cells repress cytokine production by follicular helper T cells and optimize IgG responses in mice. Eur. J. Immunol. 46, 1152–1161 (2016).

13. P. T. Sage, N. Ron-Harel, V. R. Juneja, D. R. Sen, S. Maleri, W. Sungnak, V. K. Kuchroo, W. N. Haining, N. Chevrier, M. Haigis, A. H. Sharpe, Suppression by TFR cells leads to durable and selective inhibition of B cell effector function. Nat. Immunol. 17, 1436–1446 (2016).

14. J. B. Wing, W. Ise, T. Kurosaki, S. Sakaguchi, Regulatory T cells control antigen-specific expansion of Tfh cell number and humoral immune responses via the coreceptor CTLA-4. Immunity 41, 1013–1025 (2014).

15. P. T. Sage, A. M. Paterson, S. B. Lovitch, A. H. Sharpe, The coinhibitory receptor CTLA-4 controls B cell responses by modulating T follicular helper, T follicular regulatory, and T regulatory cells. Immunity 41, 1026–1039 (2014).

16. P. T. Sage, A. H. Sharpe, T follicular regulatory cells in the regulation of B cell responses. Trends Immunol. 36, 410–418 (2015).

17. C. J. Wang, F. Heuts, V. Ovcinnikovs, L. Wardzinski, C. Bowers, E. M. Schmidt, A. Kogimtzis, R. Kenefeck, D. M. Sansom, L. S. K. Walker, CTLA-4 controls follicular helper T-cell differentiation by regulating the strength of CD28 engagement. Proc. Natl. Acad. Sci. U.S.A. 112, 524–529 (2015).

18. D. A. A. Vignali, L. W. Collison, C. J. Workman, How regulatory T cells work. Nat. Rev. Immunol. 8, 523–532 (2008).

19. B. León, J. E. Bradley, F. E. Lund, T. D. Randall, A. Ballesteros-Tato, FoxP3+ regulatory T cells promote influenza-specific Tfh responses by controlling IL-2 availability. Nat. Commun. 5, 3495 (2014).

20. J. P. Ray, M. M. Staron, J. A. Shyer, P.-C. Ho, H. D. Marshall, S. M. Gray, B. J. Laidlaw, K. Araki, R. Ahmed, S. M. Kaech, J. Craft, The interleukin-2-mTORc1 kinase axis defines the signaling, differentiation, and metabolism of T helper 1 and follicular B helper T cells. Immunity 43, 690–702 (2015).

21. Y. P. Rubtsov, J. P. Rasmussen, E. Y. Chi, J. Fontenot, L. Castelli, X. Ye, P. Treuting, L. Siewe, A. Roers, W. R. Henderson Jr., W. Muller, A. Y. Rudensky, Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 28, 546–558 (2008).

22. B. J. Laidlaw, W. Cui, R. A. Amezquita, S. M. Gray, T. Guan, Y. Lu, Y. Kobayashi, R. A. Flavell, S. H. Kleinstein, J. Craft, S. M. Kaech, Production of IL-10 by CD4+ regulatory T cells during the resolution of infection promotes the maturation of memory CD8+ T cells. Nat. Immunol. 16, 871–879 (2015).

23. B. J. Laidlaw, J. E. Craft, S. M. Kaech, The multifaceted role of CD4+ T cells in CD8+ T cell memory. Nat. Rev. Immunol. 16, 102–111 (2016).

24. F. Rousset, E. Garcia, T. Defrance, C. Péronne, N. Vezzio, D. H. Hsu, R. Kastelein, K. W. Moore, J. Banchereau, Interleukin 10 is a potent growth and differentiation factor for activated human B lymphocytes. Proc. Natl. Acad. Sci. U.S.A. 89, 1890–1893 (1992).

25. J. Choe, Y. S. Choi, IL-10 interrupts memory B cell expansion in the germinal center by inducing differentiation into plasma cells. Eur. J. Immunol. 28, 508–515 (1998).

26. S.-O. Yoon, X. Zhang, P. Berner, Y. S. Choi, IL-21 and IL-10 have redundant roles but differential capacities at different stages of plasma cell generation from human germinal center B cells. J. Leukoc. Biol. 86, 1311–1318 (2009).

27. G. Cai, X. Nie, W. Zhang, B. Wu, J. Lin, H. Wang, C. Jiang, Q. Shen, A regulatory role for IL-10 receptor signaling in development and B cell help of T follicular helper cells in mice. J. Immunol. 189, 1294–1302 (2012).

28. J. J. Guthmiller, A. C. Graham, R. A. Zander, R. L. Pope, N. S. Butler, Cutting edge: IL-10 is essential for the generation of germinal center B cell responses and anti-Plasmodium humoral immunity. J. Immunol. 198, 617–622 (2017).

29. C. L. Maynard, L. E. Harrington, K. M. Janowski, J. R. Oliver, C. L. Zindl, A. Y. Rudensky, C. T. Weaver, Regulatory T cells expressing interleukin 10 develop from Foxp3+ and Foxp3- precursor cells in the absence of interleukin 10. Nat. Immunol. 8, 931–941 (2007).

30. I. A. Parish, H. D. Marshall, M. M. Staron, P. A. Lang, A. Brüstle, J. H. Chen, W. Cui, Y.-C. Tsui, C. Perry, B. J. Laidlaw, P. S. Ohashi, C. T. Weaver, S. M. Kaech, Chronic viral infection promotes sustained Th1-derived immunoregulatory IL-10 via BLIMP-1. J. Clin. Invest. 124, 3455–3468 (2014).

31. H. D. Marshall, A. Chandele, Y. W. Jung, H. Meng, A. C. Poholek, I. A. Parish, R. Rutishauser, W. Cui, S. H. Kleinstein, J. Craft, S. M. Kaech, Differential expression of Ly6C and T-bet distinguish effector and memory Th1 CD4+ cell properties during viral infection. Immunity 35, 633–646 (2011).

32. A. C. Poholek, K. Hansen, S. G. Hernandez, D. Eto, A. Chandele, J. S. Weinstein, X. Dong, J. M. Odegard, S. M. Kaech, A. L. Dent, S. Crotty, J. Craft, In vivo regulation of Bcl6 and T follicular helper cell development. J. Immunol. 185, 313–326 (2010).

33. J. M. Odegard, B. R. Marks, L. D. DiPlacido, A. C. Poholek, D. H. Kono, C. Dong, R. A. Flavell, J. Craft, ICOS-dependent extrafollicular helper T cells elicit IgG production via IL-21 in systemic autoimmunity. J. Exp. Med. 205, 2873–2886 (2008).

34. M. A. Linterman, L. Beaton, D. Yu, R. R. Ramiscal, M. Srivastava, J. J. Hogan, N. K. Verma, M. J. Smyth, R. J. Rigby, C. G. Vinuesa, IL-21 acts directly on B cells to regulate Bcl-6 expression and germinal center responses. J. Exp. Med. 207, 353–363 (2010).

35. J. S. Hale, B. Youngblood, D. R. Latner, A. U. R. Mohammed, L. Ye, R. S. Akondy, T. Wu, S. S. Iyer, R. Ahmed, Distinct memory CD4+ T cells with commitment to T follicular helper- and T helper 1-cell lineages are generated after acute viral infection. Immunity 38, 805–817 (2013).

36. S. M. Anderson, M. M. Tomayko, A. Ahuja, A. M. Haberman, M. J. Shlomchik, New markers for murine memory B cells that define mutated and unmutated subsets. J. Exp. Med. 204, 2103–2114 (2007).

37. I. Dogan, B. Bertocci, V. Vilmont, F. Delbos, J. Mégret, S. Storck, C. A. Reynaud, J. C. Weill, Multiple layers of B cell memory with different effector functions. Nat. Immunol. 10, 1292–1299 (2009).

38. Y. Tian, S. B. Mollo, L. E. Harrington, A. J. Zajac, IL-10 regulates memory T cell development and the balance between Th1 and follicular Th cell responses during an acute viral infection. J. Immunol. 197, 1308–1321 (2016).

by guest on Septem

ber 25, 2020http://im

munology.sciencem

ag.org/D

ownloaded from

Page 10: IMMUNE REGULATION Copyright © 2017 Interleukin-10 from CD4 ...€¦ · Laidlaw et al., Sci. Immunol. 2, eaan4767 2017 20 October 2017 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 10

Laidlaw et al., Sci. Immunol. 2, eaan4767 (2017) 20 October 2017

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

10 of 10

39. Z. Shulman, A. D. Gitlin, J. S. Weinstein, B. Lainez, E. Esplugues, R. A. Flavell, J. E. Craft, M. C. Nussenzweig, Dynamic signaling by T follicular helper cells during germinal center B cell selection. Science 345, 1058–1062 (2014).

40. Y. S. Choi, R. Kageyama, D. Eto, T. C. Escobar, R. J. Johnston, L. Monticelli, C. Lao, S. Crotty, ICOS receptor instructs T follicular helper cell versus effector cell differentiation via induction of the transcriptional repressor Bcl6. Immunity 34, 932–946 (2011).

41. G. D. Victora, D. Dominguez-Sola, A. B. Holmes, S. Deroubaix, R. Dalla-Favera, M. C. Nussenzweig, Identification of human germinal center light and dark zone cells and their relationship to human B-cell lymphomas. Blood 120, 2240–2248 (2012).

42. D. Dominguez-Sola, J. Kung, A. B. Holmes, V. A. Wells, T. Mo, K. Basso, R. Dalla-Favera, The FOXO1 transcription factor instructs the germinal center dark zone program. Immunity 43, 1064–1074 (2015).

43. S. Sander, V. T. Chu, T. Yasuda, A. Franklin, R. Graf, D. P. Calado, S. Li, K. Imami, M. Selbach, M. Di Virgilio, L. Bullinger, K. Rajewsky, PI3 kinase and FOXO1 transcription factor activity differentially control B cells in the germinal center light and dark zones. Immunity 43, 1075–1086 (2015).

44. O. Bannard, Robert M. Horton, C. D. C. Allen, J. An, T. Nagasawa, J. G. Cyster, Germinal center centroblasts transition to a centrocyte phenotype according to a timed program and depend on the dark zone for effective selection. Immunity 39, 912–924 (2013).

45. T. Inoue, R. Shinnakasu, W. Ise, C. Kawai, T. Egawa, T. Kurosaki, The transcription factor Foxo1 controls germinal center B cell proliferation in response to T cell help. J. Exp. Med. 214, 1181–1198 (2017).

46. M. M. Staron, S. M. Gray, H. D. Marshall, I. A. Parish, J. H. Chen, C. J. Perry, G. Cui, M. O. Li, S. M. Kaech, The transcription factor FoxO1 sustains expression of the inhibitory receptor PD-1 and survival of antiviral CD8+ T cells during chronic infection. Immunity 41, 802–814 (2014).

47. H. White, D. Gray, Analysis of immunoglobulin (Ig) isotype diversity and IgM/D memory in the response to phenyl-oxazolone. J. Exp. Med. 191, 2209–2220 (2000).

48. J. A. Vander Heiden, P. Stathopoulos, J. Q. Zhou, L. Chen, T. J. Gilbert, C. R. Bolen, R. J. Barohn, M. M. Dimachkie, E. Ciafaloni, T. J. Broering, F. Vigneault, R. J. Nowak, S. H. Kleinstein, K. C. O’Connor, Dysregulation of B cell repertoire formation in myasthenia gravis patients revealed through deep sequencing. J. Immunol. 198, 1460–1473 (2017).

49. L. Pace, A. Tempez, C. Arnold-Schrauf, F. Lemaitre, P. Bousso, L. Fetler, T. Sparwasser, S. Amigorena, Regulatory T cells increase the avidity of primary CD8+ T cell responses and promote memory. Science 338, 532–536 (2012).

50. B. B. Ding, J. J. Yu, R. Y.-L. Yu, L. M. Mendez, R. Shaknovich, Y. Zhang, G. Cattoretti, B. H. Ye, Constitutively activated STAT3 promotes cell proliferation and survival in the activated B-cell subtype of diffuse large B-cell lymphomas. Blood 111, 1515–1523 (2008).

51. M. Gupta, J. J. Han, M. Stenson, M. Maurer, L. Wellik, G. Hu, S. Ziesmer, A. Dogan, T. E. Witzig, Elevated serum IL-10 levels in diffuse large B-cell lymphoma: A mechanism of aberrant JAK2 activation. Blood 119, 2844–2853 (2012).

52. W. Béguelin, S. Sawh, N. Chambwe, F. C. Chan, Y. Jiang, J. W. Choo, D. W. Scott, A. Chalmers, H. Geng, L. Tsikitas, W. Tam, G. Bhagat, R. D. Gascoyne, R. Shaknovich, IL10 receptor is a novel therapeutic target in DLBCLs. Leukemia 29, 1684–1694 (2015).

53. H.-Y. Chun, J.-W. Chung, H.-A. Kim, J.-M. Yun, J.-Y. Jeon, Y.-M. Ye, S.-H. Kim, H.-S. Park, C.-H. Suh, Cytokine IL-6 and IL-10 as biomarkers in systemic lupus erythematosus. J. Clin. Immunol. 27, 461–466 (2007).

54. R. J. O. Dowling, I. Topisirovic, T. Alain, M. Bidinosti, B. D. Fonseca, E. Petroulakis, X. Wang, O. Larsson, A. Selvaraj, Y. Liu, S. C. Kozma, G. Thomas, N. Sonenberg, mTORC1-mediated cell proliferation, but not cell growth, controlled by the 4E-BPs. Science 328, 1172–1176 (2010).

55. A. Roers, L. Siewe, E. Strittmatter, M. Deckert, D. Schlüter, W. Stenzel, A. D. Gruber, T. Krieg, K. Rajewsky, W. Müller, T cell-specific inactivation of the interleukin 10 gene in mice results in enhanced T cell responses but normal innate responses to lipopolysaccharide or skin irritation. J. Exp. Med. 200, 1289–1297 (2004).

56. J. M. Kim, J. P. Rasmussen, A. Y. Rudensky, Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat. Immunol. 8, 191–197 (2007).

57. N. S. Joshi, W. Cui, A. Chandele, H. K. Lee, D. R. Urso, J. Hagman, L. Gapin, S. M. Kaech, Inflammation directs memory precursor and short-lived effector CD8+ T cell fates via the graded expression of T-bet transcription factor. Immunity 27, 281–295 (2007).

58. J. P. Ray, H. D. Marshall, B. J. Laidlaw, M. M. Staron, S. M. Kaech, J. Craft, Transcription factor STAT3 and type I interferons are corepressive insulators for differentiation of follicular helper and T helper 1 cells. Immunity 40, 367–377 (2014).

59. S. Anders, D. J. McCarthy, Y. Chen, M. Okoniewski, G. K. Smyth, W. Huber, M. D. Robinson, Count-based differential expression analysis of RNA sequencing data using R and Bioconductor. Nat. Protoc. 8, 1765–1786 (2013).

60. M. G. McHeyzer-Williams, M. J. McLean, P. A. Lalor, G. J. Nossal, Antigen-driven B cell differentiation in vivo. J. Exp. Med. 178, 295–307 (1993).

61. L. Sastry, M. Alting-Mees, W. D. Huse, J. M. Short, J. A. Sorge, B. N. Hay, K. D. Janda, S. J. Benkovic, R. A. Lerner, Cloning of the immunological repertoire in Escherichia coli for generation of monoclonal catalytic antibodies: Construction of a heavy chain variable region-specific cDNA library. Proc. Natl. Acad. Sci. U.S.A. 86, 5728–5732 (1989).

62. J. Ye, N. Ma, T. L. Madden, J. M. Ostell, IgBLAST: An immunoglobulin variable domain sequence analysis tool. Nucleic Acids Res. 41, W34–W40 (2013).

63. E. Alamyar, V. Giudicelli, S. Li, P. Duroux, M.-P. Lefranc, IMGT/HighV-QUEST: The IMGT® web portal for immunoglobulin (Ig) or antibody and T cell receptor (TR) analysis from NGS high throughput and deep sequencing. Immunome Res. 8, 26 (2012).

64. N. T. Gupta, J. A. Vander Heiden, M. Uduman, D. Gadala-Maria, G. Yaari, S. H. Kleinstein, Change-O: A toolkit for analyzing large-scale B cell immunoglobulin repertoire sequencing data. Bioinformatics 31, 3356–3358 (2015).

65. G. Yaari, M. Uduman, S. H. Kleinstein, Quantifying selection in high-throughput Immunoglobulin sequencing data sets. Nucleic Acids Res. 40, e134 (2012).

66. G. Yaari, S. H. Kleinstein, Practical guidelines for B-cell receptor repertoire sequencing analysis. Genome Med. 7, 121 (2015).

67. J. N. H. Stern, G. Yaari, J. A. Vander Heiden, G. Church, W. F. Donahue, R. Q. Hintzen, A. J. Huttner, J. D. Laman, R. M. Nagra, A. Nylander, D. Pitt, S. Ramanan, B. A. Siddiqui, F. Vigneault, S. H. Kleinstein, D. A. Hafler, K. C. O’Connor, B cells populating the multiple sclerosis brain mature in the draining cervical lymph nodes. Sci. Transl. Med. 6, 248ra107 (2014).

68. M. I. Love, W. Huber, S. Anders, Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

Acknowledgments: We thank all members of the Craft laboratory for helpful discussions and critical reading of the manuscript. Funding: This work was supported by grants from the NIH [R37AR40072 (J.C.), R01AR068994 (J.C.), P30AR053495 (J.C.), R21AR063942 (J.C.), T32AI07019 (B.J.L.), F31AG07777 (B.J.L.), R01AI104739 (S.M.K.), and K01AR067892 (J.S.W.)] and the Gruber Science Fellowship (Y.L.). Author contributions: B.J.L., Y.L., S.M.K., and J.C. conceived and designed the experiments. B.J.L., Y.L., and J.S.W. performed the experiments. B.J.L., Y.L., R.A.A., J.A.V.H., N.T.G., and S.H.K. analyzed the data. B.J.L., Y.L., and J.C. wrote the manuscript. Competing interests: The authors declare that they have no competing interests. Data and materials availability: The RNA-seq and BCR sequencing data reported in this paper are archived at National Center for Biotechnology Information Gene Expression Omnibus (SRP117018).

Submitted 19 April 2017Accepted 13 September 2017Published 20 October 201710.1126/sciimmunol.aan4767

Citation: B. J. Laidlaw, Y. Lu, R. A. Amezquita, J. S. Weinstein, J. A. Vander Heiden, N. T. Gupta, S. H. Kleinstein, S. M. Kaech, J. Craft, Interleukin-10 from CD4+ follicular regulatory T cells promotes the germinal center response. Sci. Immunol. 2, eaan4767 (2017).

by guest on Septem

ber 25, 2020http://im

munology.sciencem

ag.org/D

ownloaded from

Page 11: IMMUNE REGULATION Copyright © 2017 Interleukin-10 from CD4 ...€¦ · Laidlaw et al., Sci. Immunol. 2, eaan4767 2017 20 October 2017 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 10

follicular regulatory T cells promotes the germinal center response+Interleukin-10 from CD4

Kleinstein, Susan M. Kaech and Joe CraftBrian J. Laidlaw, Yisi Lu, Robert A. Amezquita, Jason S. Weinstein, Jason A. Vander Heiden, Namita T. Gupta, Steven H.

DOI: 10.1126/sciimmunol.aan4767, eaan4767.2Sci. Immunol. 

IL-10 promoted GC B cells to adopt a dark zone phenotype.responsive and showed that−context of acute viral infection. They found dendritic cells and B cells in the GCs to be IL-10

derived IL-10 does support GC responses in the− cellfr. demonstrate that Tet al cells, Laidlaw frexpression in murine Twhether IL-10 production by these cells regulates germinal center (GC) responses in vivo. By specifically ablating IL-10

cells produce interleukin-10 (IL-10), it has been unclearfrfollicular helper T cell functions. Although it is known that T) cells express key molecules that are associated with regulatory T cell andfr follicular regulatory T (T+CD4

Probing the functions of follicular regulatory T cells

ARTICLE TOOLS http://immunology.sciencemag.org/content/2/16/eaan4767

MATERIALSSUPPLEMENTARY http://immunology.sciencemag.org/content/suppl/2017/10/17/2.16.eaan4767.DC1

REFERENCES

http://immunology.sciencemag.org/content/2/16/eaan4767#BIBLThis article cites 68 articles, 24 of which you can access for free

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Terms of ServiceUse of this article is subject to the

is a registered trademark of AAAS.Science ImmunologyNew York Avenue NW, Washington, DC 20005. The title (ISSN 2470-9468) is published by the American Association for the Advancement of Science, 1200Science Immunology

Science. No claim to original U.S. Government WorksCopyright © 2017 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of

by guest on Septem

ber 25, 2020http://im

munology.sciencem

ag.org/D

ownloaded from