Top Banner
Christoffersson et al., Sci. Immunol. 3, eaam6533 (2018) 23 March 2018 SCIENCE IMMUNOLOGY | RESEARCH ARTICLE 1 of 12 AUTOIMMUNITY Suppression of diabetes by accumulation of non–islet-specific CD8 + effector T cells in pancreatic islets Gustaf Christoffersson, 1 Grzegorz Chodaczek, 1,2 Sowbarnika S. Ratliff, 1 Ken Coppieters, 1,3 Matthias G. von Herrath 1,3 * The inflammatory lesion at the pancreatic islet in type 1 diabetes (T1D) contains a heterogeneous infiltrate of T cells. In human and mouse studies, a large majority (98 to 99%) of the cytotoxic CD8 + T cells (CTLs) within islets are not specific to any islet antigen and are thought to passively add to tissue damage. We show by intravital confocal microscopy the opposite, immune-regulatory function of this cohort of CTLs. Diabetes did not develop in mice with islets showing high levels of infiltration of non–islet-specific CTLs not recognizing local antigens. Accumulation of such CTLs resulted in lower activation and proliferation of islet-specific CTLs, leading them to enter a state of unresponsiveness due to limited access to antigens at the inflammatory lesion. This nonspecific suppression by nonautoreactive CTLs was recapitulated in a model of viral meningitis, may explain viral interference in auto- immunity, and provides insight into the regulation of organ-specific autoimmune responses. INTRODUCTION A hallmark of type 1 diabetes (T1D) is insulitis, the accumulation of an inflammatory infiltrate in the islets of Langerhans, leading to the subsequent destruction of the insulin-producing cells. The infil- trate is populated by a wide range of immune cell subsets, including the main cytotoxic effector, the CD8 + T cell. The retention of these effectors is thought to be governed by the presence of local antigen (Ag) (1), whereas their entry into solid tissues is mediated by the ac- tivation and expression of chemokines (2). Current models of the insulitic lesion in the pancreas postulate the accumulation of auto- reactive cytotoxic CD8 + T cells (CTLs). However, results from both human pathology samples (3) and mouse models (4) have shown that the level of islet Ag specificity can be as low as 1 to 2% among infil- trating CD8 + T cells, and although an autoreactive CTL clone is present in the body, it needs to reach a threshold quota of the total CTL rep- ertoire to induce T1D (4). The importance of the non–islet-specific majority of CD8 + T cells present at the islets during autoimmune dia- betes and their contribution to the course of the disease are unknown. These nonspecific CTLs are present at inflammatory lesions, areas devoid of their cognate Ag, and they have been shown to be able to add to tissue damage through the nonspecific release of cytokines without local T cell receptor (TCR) engagement (5, 6). However, it is still unclear to what extent they affect the course of an inflammatory or autoimmune disorder. The dual nature of immune cell responses adds to the com- plexity of what is occurring within the inflammatory lesion. A balance between aggressive immune responses and counteracting suppressive responses appears to be critical to avoiding excessive and unresolved inflammation. Endogenous immune suppression by the immune system has been observed for decades (7), and CD8 expression on suppressor cells was associated with this response. Failure to identify a molecular basis for the suppressive effect of these cells brought studies to a halt. The field was revived by the discovery of CD4 + /CD25 + suppressors, and since then the most studied type of suppressive immune response is that of the bona fide regulatory T cell (T reg ) (8). However, with recent insights into the regulatory mechanisms used by CD8 + T cells, the interest in the immune-suppressive effects of these cells is reawak- ening (9, 10). These, in combination with the understanding that the immune system is extremely plastic and situation-dependent, have widened the preconceptions of immune-regulatory cells to not only include the classical CD4 + T regs . In this study, we have investigated the trafficking and influence of these “bystander” CTLs not recognizing local tissue Ags on the progression to T1D in different Ag-driven mouse models of this dis- ease and one model of viral meningitis. We found that activated CTLs not recognizing local tissue Ags can elicit powerful suppressive effects when present in inflammatory lesions by limiting the local access to target cells and Ag-presenting cells for the target-specific effectors and, thereby, acted protectively against disease. RESULTS Ag governs CTL behavior at islets, and inflammation is required for their accumulation in larger numbers T cells can gain access to sites of inflammation even though their cog- nate Ag is not expressed at that location, which is evident at the pan- creatic islets in T1D (3, 4). We decided to investigate the requirements for the accumulation of non–islet-specific CD8 + T cells at pancreatic islets in well-defined Ag-driven models of autoimmune diabetes. Using mice expressing the lymphocytic choriomeningitis virus (LCMV) glycoprotein (GP) under the control of the rat insulin promoter (RIP), we adoptively transferred TCR-transgenic (TCR-tg) GP-specific P14 CD8 + T cells (fig. S1A). Transferred naïve P14 cells did not accumu- late in the pancreas of RIP-GP mice but required stimulation through LCMV infection of the host or by GP 33–41 (KAVYNFATC) peptide immunization to enter the islets and destroy the cells (fig. S2). In the face of ongoing insulitis, we also investigated the trafficking of non–islet-specific OT-I CD8 + T cells to the islets. These cells are spe- cific for chicken egg ovalbumin (OVA), do not recognize Ag in the RIP-GP mouse, and did not accumulate in the insulitic lesions in their naïve state. However, when they were activated by either recombinant LCMV-OVA virus (fig. S1A) or OVA 257–264 (SIINFEKL) immuni- zation (fig. S1B), they infiltrated and accumulated at the inflamed islets to a similar extent to and in the same pattern as their activated 1 Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA. 2 Wroclaw Research Centre EIT+, Wroclaw, Poland. 3 Novo Nordisk Diabetes Research and Development Center, Seattle, WA 98109, USA. *Corresponding author. Email: [email protected] Copyright © 2018 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 March 26, 2021 http://immunology.sciencemag.org/ Downloaded from
13

AUTOIMMUNITY Copyright © 2018 Suppression of diabetes by ...€¦ · Christoffersson et al., Sci. Immunol. 3, eaam6533 2018 23 March 2018 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of

Oct 19, 2020

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: AUTOIMMUNITY Copyright © 2018 Suppression of diabetes by ...€¦ · Christoffersson et al., Sci. Immunol. 3, eaam6533 2018 23 March 2018 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of

Christoffersson et al., Sci. Immunol. 3, eaam6533 (2018) 23 March 2018

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 12

A U T O I M M U N I T Y

Suppression of diabetes by accumulation of non–islet-specific CD8+ effector T cells in pancreatic isletsGustaf Christoffersson,1 Grzegorz Chodaczek,1,2 Sowbarnika S. Ratliff,1 Ken Coppieters,1,3 Matthias G. von Herrath1,3*

The inflammatory lesion at the pancreatic islet in type 1 diabetes (T1D) contains a heterogeneous infiltrate of T cells. In human and mouse studies, a large majority (98 to 99%) of the cytotoxic CD8+ T cells (CTLs) within islets are not specific to any islet antigen and are thought to passively add to tissue damage. We show by intravital confocal microscopy the opposite, immune-regulatory function of this cohort of CTLs. Diabetes did not develop in mice with islets showing high levels of infiltration of non–islet-specific CTLs not recognizing local antigens. Accumulation of such CTLs resulted in lower activation and proliferation of islet-specific CTLs, leading them to enter a state of unresponsiveness due to limited access to antigens at the inflammatory lesion. This nonspecific suppression by nonautoreactive CTLs was recapitulated in a model of viral meningitis, may explain viral interference in auto-immunity, and provides insight into the regulation of organ-specific autoimmune responses.

INTRODUCTIONA hallmark of type 1 diabetes (T1D) is insulitis, the accumulation of an inflammatory infiltrate in the islets of Langerhans, leading to the subsequent destruction of the insulin-producing cells. The infil-trate is populated by a wide range of immune cell subsets, including the main cytotoxic effector, the CD8+ T cell. The retention of these effectors is thought to be governed by the presence of local antigen (Ag) (1), whereas their entry into solid tissues is mediated by the ac-tivation and expression of chemokines (2). Current models of the insulitic lesion in the pancreas postulate the accumulation of auto-reactive cytotoxic CD8+ T cells (CTLs). However, results from both human pathology samples (3) and mouse models (4) have shown that the level of islet Ag specificity can be as low as 1 to 2% among infil-trating CD8+ T cells, and although an autoreactive CTL clone is present in the body, it needs to reach a threshold quota of the total CTL rep-ertoire to induce T1D (4). The importance of the non–islet-specific majority of CD8+ T cells present at the islets during autoimmune dia-betes and their contribution to the course of the disease are unknown.

These nonspecific CTLs are present at inflammatory lesions, areas devoid of their cognate Ag, and they have been shown to be able to add to tissue damage through the nonspecific release of cytokines without local T cell receptor (TCR) engagement (5, 6). However, it is still unclear to what extent they affect the course of an inflammatory or autoimmune disorder. The dual nature of immune cell responses adds to the com-plexity of what is occurring within the inflammatory lesion. A balance between aggressive immune responses and counteracting suppressive responses appears to be critical to avoiding excessive and unresolved inflammation. Endogenous immune suppression by the immune system has been observed for decades (7), and CD8 expression on suppressor cells was associated with this response. Failure to identify a molecular basis for the suppressive effect of these cells brought studies to a halt. The field was revived by the discovery of CD4+/CD25+ suppressors, and since then the most studied type of suppressive immune response is that of the bona fide regulatory T cell (Treg) (8). However, with recent

insights into the regulatory mechanisms used by CD8+ T cells, the interest in the immune-suppressive effects of these cells is reawak-ening (9, 10). These, in combination with the understanding that the immune system is extremely plastic and situation-dependent, have widened the preconceptions of immune-regulatory cells to not only include the classical CD4+ Tregs.

In this study, we have investigated the trafficking and influence of these “bystander” CTLs not recognizing local tissue Ags on the progression to T1D in different Ag-driven mouse models of this dis-ease and one model of viral meningitis. We found that activated CTLs not recognizing local tissue Ags can elicit powerful suppressive effects when present in inflammatory lesions by limiting the local access to target cells and Ag-presenting cells for the target-specific effectors and, thereby, acted protectively against disease.

RESULTSAg governs CTL behavior at islets, and inflammation is required for their accumulation in larger numbersT cells can gain access to sites of inflammation even though their cog-nate Ag is not expressed at that location, which is evident at the pan-creatic islets in T1D (3, 4). We decided to investigate the requirements for the accumulation of non–islet-specific CD8+ T cells at pancreatic islets in well-defined Ag-driven models of autoimmune diabetes. Using mice expressing the lymphocytic choriomeningitis virus (LCMV) glycoprotein (GP) under the control of the rat insulin promoter (RIP), we adoptively transferred TCR-transgenic (TCR-tg) GP-specific P14 CD8+ T cells (fig. S1A). Transferred naïve P14 cells did not accumu-late in the pancreas of RIP-GP mice but required stimulation through LCMV infection of the host or by GP33–41 (KAVYNFATC) peptide immunization to enter the islets and destroy the cells (fig. S2). In the face of ongoing insulitis, we also investigated the trafficking of non–islet-specific OT-I CD8+ T cells to the islets. These cells are spe-cific for chicken egg ovalbumin (OVA), do not recognize Ag in the RIP-GP mouse, and did not accumulate in the insulitic lesions in their naïve state. However, when they were activated by either recombinant LCMV-OVA virus (fig. S1A) or OVA257–264 (SIINFEKL) immuni-zation (fig. S1B), they infiltrated and accumulated at the inflamed islets to a similar extent to and in the same pattern as their activated

1Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA. 2Wroclaw Research Centre EIT+, Wroclaw, Poland. 3Novo Nordisk Diabetes Research and Development Center, Seattle, WA 98109, USA.*Corresponding author. Email: [email protected]

Copyright © 2018 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 March 26, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 2: AUTOIMMUNITY Copyright © 2018 Suppression of diabetes by ...€¦ · Christoffersson et al., Sci. Immunol. 3, eaam6533 2018 23 March 2018 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of

Christoffersson et al., Sci. Immunol. 3, eaam6533 (2018) 23 March 2018

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 12

islet-specific P14 counterparts (Fig. 1A and movie S1). Simultaneous islet inflammation and the expression of an auto-Ag were require-ments for the accumulation of both activated P14 and activated OT-I CD8+ T cells because, in wild-type (wt) C57Bl/6J mice, where neither the GP nor the OVA Ag is expressed on the islet cells, no accumu-lation of transferred and activated T cells was observed (Fig. 1B and movie S2). The expansion of the transferred T cells was comparable between the wt and RIP-GP strains, as measured by their numbers in the spleen (Fig. 1, C and D). In groups where both transferred P14 and OT-I CD8+ T cells were activated and GP Ag was expressed on cells, both populations of CTLs were found to specifically accumu-late at the islets and not dwell in the exocrine parenchyma to any larger extent (Fig. 1, E to G). This highly focal accumulation of non–islet-specific CTLs observed at the islets was unexpected. This was also found to occur to the same extent in the RIP-mOVA mouse model where the antigenic situation is reversed (OVA is expressed on the cells; fig. S1C): Non–islet-specific P14 CTLs accumulated at the islets in the presence of islet-specific OT-I CTLs. These data indi-cate that, irrespective of the model (RIP-GP or RIP-mOVA), the trans-ferred non–islet-specific CTLs were found to have the same tissue distribution patterns as their islet-specific counterparts despite the com-plete lack of cognate Ag in the pancreas.

We further analyzed the behavior of the two cell populations at the islets using intravital confocal microscopy of the pancreas (Fig. 1H) (11). Imaging was performed 7 to 9 days after adoptive transfer of TCR transgenic CD8+ T cells, when islet inflammation is at its peak in these models (Fig. 1I). Tracking the migration of the transferred CTLs re-vealed that islet-specific CTLs were moving substantially slower than their Ag-ignorant counterparts (Fig. 1, J and K). We also found that the non–islet-specific cells were, on average, more distant to the center of the islet, which was mostly occupied by the islet-specific CTLs (Fig. 1L). This migration pattern points to the notion that although Ag was dispensable for the accumulation of non–islet-specific CTLs at in-flammatory foci, it governed the behavior of the CTLs in the micro-environment of the islet.

Abundance of activated non–islet-specific CTLs at islets is immune-suppressiveWe found the OT-I transgenic CD8+ T cells to differ in their activation kinetics to their cognate Ag compared with the CD8+ T cells of the P14 transgene [also observed by others (12)]. The OT-I TCR-tg cells ex-panded several logs faster than the P14 TCR-tg in similar induction protocols (LCMV-OVA infection or SIINFEKL + KAVYNFATC im-munization). This required optimization of immunization and adop-tive transfer protocols to have quantifiable amounts of cells at the islets for imaging purposes. During the process of finding an ap-propriate balance between the numbers of the two TCR transgenes, we found that the transfer of equally high amounts (1 × 106 cells; ratio, 1:1) of P14 and OT-I CD8+ T cells (a scenario hereon denoted “HI”) to the RIP-GP model did not result in cell death and hyper-glycemia (Fig. 2, A to E) despite undergoing an intensive immuniza-tion protocol (fig. S1B). In these animals, we observed massive insulitis at days 7 to 9 after adoptive transfer (Fig. 2A) and the local accumula-tion of both P14 and OT-I CTLs, with particularly high amounts of the latter. However, when imaging the same group of animals 1 week later, both types of CTLs had retracted from the pancreas (Fig. 2B). Islets were mostly intact, and six of eight mice were normoglycemic (Fig. 2F). We then lowered the amount of transferred OT-I CD8+ T cells in logarithmic decrements and found that when 1 × 104 (1:100) or 1 ×

103 (1:1000) cells were cotransferred with 1 × 106 P14 CD8+ T cells, diabetes incidence again became normal for this model (95 to 100%; Fig. 2E) (13). In contrast to the observations made in the HI scenario, mice that had received only 1 × 103 (1:1000) non–islet-specific OT-I CD8+ T cells (a scenario hereon denoted “LO”) also presented with insulitis on days 7 to 9, along with signs of cell destruction (Fig. 2C). One week later, these animals were overtly diabetic (seven of eight mice were hyperglycemic; Fig. 2F), and very few cells remained intact in these pancreata (Fig. 2D). In addition, in this scenario, the CTLs had largely retracted from the pancreas. Similar observations were made when animals, instead of being immunized by peptides, were infected with the recombinant LCMV-OVA virus to activate both TCR trans-genes (fig. S1A).

To rule out the fact that the observations made in the RIP-GP mouse were not model-specific, we reversed the antigenic situation in the RIP-mOVA mouse (fig. S1C). We adjusted the numbers of transferred CTLs to accommodate the differences in antigenic reac-tivity between the P14 and OT-I transgenes. We found that 1 × 104 transferred OT-I CD8+ effectors in the RIP-mOVA model resulted in diabetes induction kinetics largely equivalent to that of 1 × 106 transferred P14 CD8+ T effectors in the RIP-GP model. Transfer-ring 2 × 106 P14 CD8+ T cells (HI) alongside the OT-I CD8+ T cells in the RIP-mOVA model mimicked the protection from diabetes seen in the RIP-GP model (five of six mice were normoglycemic; Fig. 2G). A similar pattern (three of four mice were normoglycemic) was also seen in the RIP-OVAlow model (fig. S1D), where Ag is expressed at very low levels on the cells and Ag is not cross-presented in pan-creatic lymph nodes (Fig. 2H).

To rule out the possibility of adoptive transfer bias (12) or the idea that an abnormal accumulation of also islet-specific CTLs at the islets could have suppressive effects, we made single transfers of 2 × 106 and 1 × 107 P14 CD8+ T cells in the RIP-GP model. These mice displayed normal amounts of P14 CTLs in insulitic lesions and had similar diabetes induction kinetics to the normal scenario when 1 × 106 P14 CD8+ T cells were transferred (fig. S3). We were also in-terested to see whether an Ag-experienced but not highly activated non–islet-specific T cell population could home to the pancreas and induce protection. We therefore induced a memory phenotype of the OT-I CD8+ T cells in vitro by culturing the cells in the presence of SIINFEKL and interleukin-7 (IL-7). In contrast to their CTL counter-parts, these cells did not accumulate in the islets or confer any pro-tection against autoimmunity after transfer to the RIP-GP model, underlining the observation that a high level of activation of CTLs is required for the migration to the islets and the nonspecific interfer-ence with an Ag-driven immune response.

Protection from autoimmunity is not due to the expansion of Treg subsetsWe next looked into the distribution of immune cells in the pancreas of the RIP-GP mouse during the HI and LO scenarios of adoptive transfer and induction of autoimmunity. By flow cytometry, the CD4+ T cell populations were similar in numbers between the groups, whereas, as expected, the CD8+ T cells were three times more abundant in the mice that had received high amounts of OT-I CD8+ T cells (Fig. 3, A to D). However, the P14 CD8+ T cells were significantly less frequent in the mice that had received high (HI) amounts of non–islet-specific OT-I CD8+ T cells (1 × 106 cells), despite receiving the same amount of islet-specific driver T cells (1 × 106 cells) as the LO group (Fig. 3E). Although this dimidiation of the amount of P14 CD8+

by guest on March 26, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 3: AUTOIMMUNITY Copyright © 2018 Suppression of diabetes by ...€¦ · Christoffersson et al., Sci. Immunol. 3, eaam6533 2018 23 March 2018 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of

Christoffersson et al., Sci. Immunol. 3, eaam6533 (2018) 23 March 2018

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 12

P14 OT-I P14 OT-I0.000

0.002

0.004

0.006

2 )

Endocrine Exocrine

0

50

100

150

200

250

0 200 400 600 800 1000 1200

Sig

nal i

nten

sity

(ar

bitra

ry u

nits

)

Distance (µm)

P14

OT-I

ExocrineExocrineIslet Islet

*

*A

B

E

F G

D

C

P14 OT-I Laser reflection

0 5 10 15 20 2560

70

80

90

100

110

Time (min)

Dis

tanc

eto

isle

tcen

ter(

µm)

P14 (Ag-spec)OT-I (not rec Ag)

*

P14(A

g-spe

c)

(not re

c Ag)

0.00

0.05

0.10

0.15

0.20

Mig

ratio

nsp

eed

(µm

/s)

*

H

K

JI

L

P14 OT-I Laser reflection P14 tracks OT-I tracks Laser reflection

µ

Fig. 1. Ag governs CTL behavior at islets but is not required for their accumulation in large numbers. (A) Islet (dashed line) in the pancreas of a diabetic RIP-GP mouse where islet-specific P14 CTLs (red) and non–islet-specific OT-I CTLs (green) have gathered. (B) An islet (dashed line) in the pancreas of a wt C57Bl/6 mouse immu-nized in the same way as in (A) displaying a few CTLs in the pancreatic parenchyma. (C and D) The levels of T cell proliferation were similar between the two strains of mice, as judged by T cell content in spleens. (E to G) The densities of accumulated CTLs of both specificities (P14 and OT-I) were higher in the islets than in the surrounding pancreatic parenchyma, as quantified in (E) (n = 5 mice) and visualized in (F) and (G), where the yellow line across a part of the pancreas in (F) is represented in the histograms over fluorescent signals in (G). (H) Islets in mouse pancreata were imaged in anesthetized mice using a vacuum imaging window where the pancreas was immobilized, enabling long-term imaging. (I and J) A still frame (z-projection) (I) from an intravital recording of an inflamed islet in a RIP-GP mouse (red P14 CD8+ T cells and green OT-I CD8+ T cells) and the resulting T cell tracks (J). (B) and (C) correspond to movie S3. (K and L) OT-I CTLs consistently displayed higher migration speeds than the islet-specific P14 CTLs (K) and were kept, on average, further away from the islet center (L). Data in (J) to (L) are from one of six representative experiments. Data are means ± SEM. *P < 0.05, two-tailed unpaired Mann-Whitney U tests. Scale bars, 20 m (A to D), 30 m (F), and 50 m (I and J).

by guest on March 26, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 4: AUTOIMMUNITY Copyright © 2018 Suppression of diabetes by ...€¦ · Christoffersson et al., Sci. Immunol. 3, eaam6533 2018 23 March 2018 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of

Christoffersson et al., Sci. Immunol. 3, eaam6533 (2018) 23 March 2018

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 12

T cells in the pancreas might have contributed to a slower progression of the autoimmune process, the amount of islet-specific cells pres-ent at the islets in these mice should be more than enough to cause cell demise and hyperglycemia. Looking into the distribution of the TCR transgenes and the endogenous CD8+ T cell populations in the pancreas, the high abundance of OT-I CD8+ T cells in the HI scenario left little space for other populations (~13%) compared with the transferred ones, whereas in the LO scenario, the space was taken up to ~87% by endogenous CD8+ T cells (Fig. 3, F to I).

An explanation to the protection from autoimmunity observed in the HI scenario could be the permissive expansion of a Treg pop-ulation. We did not find any significant differences in the CD4+/CD25+/FoxP3+ subset of regulatory cells in the pancreas (Fig. 3, J to L) or pancreatic draining lymph nodes between the groups to explain the immune suppression. Markers for regulatory-like CD8+ T cells, such as FoxP3 (Fig. 3M) or CD122 (Fig. 3N) (14), did not show any differences in numbers or levels of expression between the HI and

LO scenarios. Thus, differences in Treg subsets did not seem to be the fac-tor limiting the expansion and actions of the islet-specific CD8+ T cells.

Accumulation of CTLs not recognizing local tissue Ag attenuates effector functions of islet-specific CTLsWe extended the profiling of the CD8+ T cells present in the pancreas to assess markers for their activation status, effector functions, and exhaustion (Fig. 4A). The non–islet-specific OT-I CD8+ T cells did not display any significant differences between the two scenarios. A few markers stood out for the islet-specific CD8+ T cells: P14 T cells in the HI scenario expressed significantly less CD69 (Fig. 4, B and C), con-sistent with their inability to execute their effector functions and kill Ag-expressing cells (15). PD-1, an inhibitory receptor that is highly expressed on CD8+ T cells after exhaustion (16), was up-regulated on islet-specific cells in the HI scenario (Fig. 4, D and E). However, other hall-mark inhibitory receptors for exhaustion [lymphocyte-activation gene 3 (LAG-3) and T cell immunoglobulin and mucin-domain containing-3

0 5 10 150

50

100

Time (days)

Non

diab

etic

anim

als

(%)

1:110:1100:11000:1

RIP-GP RIP-OVAlowRIP-mOVA

HI (1:1) LO (1000:1)

9 yaD

61 yaD

E

HGF

A

B D

C

P14 OT-I Laser reflection

0 5 10 150

200

400

600

Time (days)

Bloo

d gl

ucos

e co

ncen

tratio

n (m

g/dl

)

HILO

0 5 10 150

200

400

600

Time (days)

Bloo

d gl

ucos

e co

ncen

tratio

n (m

g/dl

)

HILO

0 5 10 150

200

400

600

Time (days)

Bloo

d gl

ucos

e co

ncen

tratio

n (m

g/dl

)

HILO

*

Fig. 2. Abundance of non–islet-specific CTLs at islets is immune-suppressive. (A and C) Adoptive transfer of equal amounts (1 × 106) of P14 and OT-I CD8+ T cells re-sulted in an abundance of non–islet-specific OT-I CTLs at islets on day 9 in RIP-GP mice (A) compared with when the ratio was changed to 1000:1 (P14:OT-I) (C). (B and D) Follow-up studies of the mice on day 16 showed that the infiltrate was gone, and in the HI (1:1 ratio) group, the islets were found to be mostly intact (B), whereas in the LO (1000:1 ratio) group, very few cells were found (D). Scale bars, 30 m. (E) A majority of the HI group (six of eight) displayed normoglycemia, whereas only one of eight was normoglycemic in the LO group (*P = 0.0058, 1:1 versus 1:1000, log-rank test). Different ratios of cell transfers suggested a dose-dependent mechanism because 50% of the mice in the group receiving a 10:1 ratio remained normoglycemic. (F to H) These scenarios were repeated in three different transgenic models that express model Ags on cells: RIP-GP (six of eight were normoglycemic, representative of four independent experiments) (F), RIP-mOVA (five of six were normoglycemic, representative of two independent experiments) (G), and RIP-OVAlow (three of four were normoglycemic, representative of two independent experiments) (H).

by guest on March 26, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 5: AUTOIMMUNITY Copyright © 2018 Suppression of diabetes by ...€¦ · Christoffersson et al., Sci. Immunol. 3, eaam6533 2018 23 March 2018 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of

Christoffersson et al., Sci. Immunol. 3, eaam6533 (2018) 23 March 2018

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 12

CD410.6

CD85.86

0–103

103

104

105

Comp-BV 570-A :: CD4

0

–103

103

104

105

Com

p-B

V 6

50-A

:: C

D8

CD46.00

CD836.9

0–103

103

104

105

Comp-BV 570-A :: CD4

0

–103

103

104

105

Com

p-B

V 6

50-A

:: C

D8

HI (1:1) LO (1000:1)*

*

HI (1:1) LO (1000:1)

HI (1:1) LO (1000:1)

A B C D

GE F

M

LKJ

I

H

P14

OT-

I non

-TC

R-tg

Las

er re

flect

ion

HI LO0

20

40

60

80

P14

T ce

lls in

pan

crea

s(%

of n

on–O

T-I C

D8+ T

cel

ls)

HI (

1:1)

LO (1

000:

1)

N

CD4

CD

8

CD4

CD

8

CD25

FoxP

3

CD25

FoxP

3

HI LO0

100

200

300

Expr

essi

on le

vel o

f C

D12

2 on

OT-

I CD

8+ T

cel

ls (M

FI)

HI LO0

2

4

6

8

10

T reg

s in

isle

ts(%

CD

4+ /C

D25

+ /Fo

xP3+

T ce

lls)

HI LO0.0

0.1

0.2

0.3

0.4

OT-

I CD

8+ T r

egs i

n is

lets

(%O

T-I

CD

8+ /Fo

xP3+

T c

ells

)

HI LO0

10

20

30

40

50

CD

8+T

cells

inpa

ncre

as(%

ofto

tall

ymph

ocyt

es)

HI LO0

5

10

15

20

CD

4+T

cells

inpa

ncre

as(%

ofno

n-C

D8+

lym

phoc

ytes

)

P14OT-Inon-TCRtg

Fig. 3. Protection from autoimmunity is not due to the expansion of Treg subsets. (A to D) Flow plots showing similar representation of CD4+ T cells at islets in the two scenarios (A and B) but different amounts of CD8+ T cells (C and D). (E) Despite having lower amounts of total CD8+ T cells, LO mice had more islet-specific P14 T cells in the pancreas. (F and G) The dis-tributions of Ag specificity of the CD8+ T cells present in the pancreas are visualized in (F) and (G), where each dot represents 1of 100. (H and I) Rep-resentative fixed, frozen tissue sections from HI and LO pancreata displaying the TCR-tg cells [red, P14 (islet-specific); green, OT-I (non–islet-specific)] and endogenous CD8+ T cells [blue, anti-CD8 monoclonal antibody (mAb) (polyclonal)] in islets (dashed lines). Scale bar, 20 m. (J to L) CD4+/CD25+/FoxP3+ Treg fractions were not different between the two scenarios. (M and N) The abundant population of non–islet-specific T cells in the HI scenario had not transformed into a CD8+ Treg type as judged by the expression of FoxP3 (M) and CD122 (N). Groups are representative of at least three inde-pendent experiments. Data are means ± SEM. *P < 0.05, two-tailed unpaired Mann-Whitney U tests. MFI, mean fluorescence intensity.

by guest on March 26, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 6: AUTOIMMUNITY Copyright © 2018 Suppression of diabetes by ...€¦ · Christoffersson et al., Sci. Immunol. 3, eaam6533 2018 23 March 2018 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of

Christoffersson et al., Sci. Immunol. 3, eaam6533 (2018) 23 March 2018

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 12

(TIM-3)] did not differ (Fig. 4A), although the decreased killer cell lectin-like receptor subfamily G member 1 (KLRG1) expression on islet-specific CD8+ T cells also points to an unresponsive phenotype (16).

To further determine the potential functional differences in the CTLs, we isolated T cells from the pancreatic draining lymph nodes of both HI and LO RIP-GP mice and stimulated them in vitro with KAVYNFATC (cognate Ag for P14) or SIINFEKL (cognate Ag for OT-I). Subsequent intracellular cytokine staining revealed that the P14 CD8+ T cells from the HI scenario produced lower amounts of interferon- (IFN-) (Fig. 5, C and D) and higher amounts of IL-10 (Fig. 5, E and F) than their counterparts in the LO scenario (Fig. 5G). We found no differences between the OT-I CD8+ T cells from the two scenarios. The apparent down-regulation of effector functions and the increased production of IL-10 in the islet-specific cells are symp-tomatic of exhausted T cells (16).

We further hypothesized that the high amount of CD8+ T cells that had gathered at the inflamed islets in the HI scenario could passively deplete the organ of inflammation-supportive and proliferative cyto-kines. To this end, we assessed the pancreatic content of 10 different cytokines to see whether increased CTL content could shift the envi-ronment in an organ to a less permissive and more immune-suppressive state. None of the assessed cytokines was differentially expressed be-tween the HI and LO scenarios when assessing the whole pancreas (fig. S4) speaking against a depletion mechanism.

Low accessibility to Ag and professional Ag-presenting cells for effector CTLs leads to reduced proliferation of effectorsWe sought to find the underlying reason for the P14 CD8+ T cells to display a nonresponsive phenotype (Fig. 4). Exhaustion occurs in sit-uations such as chronic viral infections, malignancies, and autoimmu-nity where CTLs are exposed to high amounts of Ag for extended periods of time, while they are not capable of completing their task (i.e., target cell killing). In the current study, the P14 CD8+ T cells in RIP-GP animals were initially exposed to high amounts of Ag (1 + 1 mg of KAVYNFATC) and adjuvant (50 + 50 g of CpG) during the im-munization protocol (fig. S1B). What was apparent in the HI scenario was that the islet-specific T cells did not fully execute their effector functions and left most cells unharmed. We performed intravital imaging in RIP-GPxCX3CR1+/GFP mice undergoing the HI and LO scenarios. Professional antigen-presenting cells (APCs) in the islets expressed green fluorescent protein (GFP) in this model and thereby enabled interrogation of the interactions between CTLs and APCs. In the in vivo recordings, it was evident that interactions between islet- specific CD8+ T cells and APCs were few in the HI scenario compared with the LO scenario. Although islet-specific P14 CD8+ T cells were fewer at the islets in the HI scenario (Fig. 3E), they were fractionally less engaged to APC than the islet-specific CD8+ T cells in the LO scenario (28 ± 17% versus 74 ± 11% occupancy, respectively; Fig. 5, A to C, and movies S4 and S5). By performing anti-CD8 immuno-fluorescence stainings of fixed, frozen pancreata, we could assess the APC occupancy by other CD8+ T cells. In the HI scenario, non–islet- specific T cells were found to occupy a majority of the APCs in the islets (61%; Fig. 5D) compared with the LO scenario where only 7% of the APCs were occupied by non–islet-specific CD8+ T cells (Fig. 5E). In both scenarios, a majority of the islet APCs were found to interact with CD8+ T cells, but a slightly larger proportion of the APCs in the LO scenario were unoccupied (HI versus LO, 8% versus 18%, respectively).

The phenotype and accumulation of APCs in islets can have a major impact on disease progression (17, 18). We therefore isolated

islet APCs from pancreata from the two scenarios and performed flow cytometry, looking for differences in three major macrophage/dendritic cell (DC) subsets—resident macrophages (Fig. 5F), migra-tory DCs (Fig. 5G), and plasmacytoid DCs (Fig. 5H)—without find-ing any differences in these compartments.

The importance of cross-presentation of Ag to CD8+ T cells at the islets is unclear, but the T cell proliferative effect of Ag presenta-tion at nonlymphatic peripheral sites of inflammation is becoming increasingly appreciated (19–22). We therefore investigated whether the apparent blocking of CTL-APC interactions that occurred in the HI scenario affected the proliferation of islet-specific CD8+ T cells. By transferring 1 × 106 CellTrace Violet–loaded CD45.1+ P14 CD8+ T cells on day 6, we were able to track the proliferation of islet-specific cells during the onset of disease by assessing the dilution of dye on days 8 and 9 in pancreatic draining lymph nodes (Fig. 5, I and J). Conforming to the findings on the amount of P14 CD8+ T cells and their activation in the HI scenario (Fig. 3E), these cells also seemed to be restricted in their proliferation. Thus, the high amount of non–islet-specific CD8+ T cells at the insulitic lesion did not seem to be detrimental to the cells but rather suppressed the autoreactive re-sponse partly by interfering with local Ag presentation, a conclusion that was further supported by the suppression observed in the RIP- OVAlow model (Fig. 2H), where islet Ag is not cross-presented in lymph nodes.

Non–viral Ag–specific CTLs dampen inflammation in a mouse model of viral meningitisIntracerebral challenge of mice with LCMV results in infection of the meninges, leptomeninges, ependyma, and choroid plexus cells. Immunocompetent mice normally succumb to the infection about 7 days after inoculation due to excessive inflammation leading to edema and brain herniation (23). To explore the wider relevance of non–Ag-specific immune suppression, we infected mice intracere-brally with LCMV after receiving either 1 × 103 or 1 × 106 activated OT-I CD8+ T cells and followed their disease progression. Mice with induced memory to LCMV through previous intraperitoneal infec-tion with the virus did not show any signs of disease, whereas mice receiving no cell transfers had a median survival of 7.25 days after intracerebral infection, as expected (Fig. 6A). The group of mice re-ceiving 1 × 103 OT-I CD8+ T cells (corresponding to the LO groups in the T1D models) mimicked the disease kinetics of the group re-ceiving no transfer and had a median survival of 7.5 days. The group of mice receiving a high amount of OT-I CD8+ T cells (1 × 106 cells corresponding to the HI groups in the T1D models) survived, on av-erage, 3 days longer, with a median survival of 10.5 days, with no ex-tended period of being moribund (Fig. 6A).

In a separate experiment, we sacrificed animals from the two groups receiving different amounts of OT-I CD8+ T cells on day 7 after infection to evaluate the cerebral inflammation. In hematoxylin and eosin stainings of the brains, we could not observe any major dis-similarity in morphology or overall cellular infiltration between the groups (Fig. 6, B and C). When staining for CD8 in fixed, frozen brain sections, we found accumulation of lymphocytes in periven-tricular regions to the same extent in both groups (Fig. 6, D and E). However, when looking for GFP+ OT-I T cells, we found differences first in the accumulation of OT-I cells, which were more or less ab-sent in the groups receiving 1 × 103 OT-I, but constituted more than 50% of the CD8+ lymphocytic infiltrate in the group receiving 1 × 106 OT-I (Fig. 6F).

by guest on March 26, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 7: AUTOIMMUNITY Copyright © 2018 Suppression of diabetes by ...€¦ · Christoffersson et al., Sci. Immunol. 3, eaam6533 2018 23 March 2018 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of

Christoffersson et al., Sci. Immunol. 3, eaam6533 (2018) 23 March 2018

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 12

IFN-γ IL-2 IL-10 TNF-α0

20

40

60

CD

8+

T c

ells

exp

ress

ing

cyto

kine

(%

)

HILO

*

*

HI (1:1) LO (1000:1)

IFN-γFSC

HI LO0

20

40

60

Expr

essi

on le

vel o

f CD

69 o

nP1

4 C

D8+

T c

ells

(MFI

)

HILOIsotype

CD69

HILOIsotype

HI LO0

50

100

150

200

Expr

essi

on le

vel o

f PD

-1 o

nP1

4 C

D8+

T c

ells

(MFI

)

*

*

PD-1

H

HI LO

CD122

CD25

CD44

CD62L

CD69

CD8

CTLA-4

FoxP3

KLRG1

LAG-3

PD-1

BA C

E

GF

I

D

J

Fig. 4. High numbers of non–islet-specific CTLs attenuate effector functions of islet-specific CTLs. (A) Flow cytometric analysis of a range of surface markers on islet-specific P14 CD8+ T cells visualized as a heat map [based on arbitrary intensity units (MFI)]. Significant differences were found in CD69, KLRG1, and PD-1. (B and C) The expression of the activation marker CD69 on Ag-specific P14 CTLs was higher in the group receiving low amounts of non–islet-specific CD8+ T cells. (D and E) The exhaus-tion marker PD-1 was found to be highly expressed in the HI scenario (D), quantified in (E). Results are representative of two to three independent experiments. Data are means ± SEM. *P < 0.05, two-tailed unpaired Mann-Whitney U tests. (F to J) Islet-specific P14 CTLs isolated from pancreatic draining lymph nodes were stimulated in vitro with their cognate Ag GP33–41 and revealed less effector cytokine production (IFN-) [(F) and (G)] and increased IL-10 production [(H) and (I)] in mice receiving HI amounts of non–islet-specific TCR-tg CD8+ T cells; this is quantified in (J). *P < 0.05, two-tailed unpaired Mann-Whitney U tests. FSC, forward scatter.

by guest on March 26, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 8: AUTOIMMUNITY Copyright © 2018 Suppression of diabetes by ...€¦ · Christoffersson et al., Sci. Immunol. 3, eaam6533 2018 23 March 2018 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of

Christoffersson et al., Sci. Immunol. 3, eaam6533 (2018) 23 March 2018

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 12

0-103

103

104

105

Comp-CellTrace Violet-A :: proliferation

0

20

40

60

80

100

Nor

mal

ized

To

Mod

e

HI LO0

20

40

60

80

100

Pro

lifer

atin

g C

D8+

P14

T c

ells

(%)

*

Proliferated

CellTrade Violet

islet-spec cells

Islet-specNon–islet-specNot occupied

A B C

D

JI

P14 CX3CR1 Laser reflection P14 CX3CR1 Laser reflection

P14 CX3CR1 OT-I Laser reflection P14 CX3CR1 OT-I Laser reflection

HILO

)1:0001( OL)1:1( IH

HI (1:1) LO (1000:1)

HI LO

0

20

40

60

80

100

Arr

est c

oeffi

cien

t (%

)

*

E

HI LO0

2

4

6

8

10

Isle

t res

iden

t mac

roph

ages

(%F4

/80+ /C

D11

b+ /CD

11c+ c

ells

of

CD

45+ /C

D19

– /MH

CII+ /F

4/80

+ /CD

11b+ )

HI LO0.0

0.5

1.0

1.5

2.0

Isle

t mig

rato

ry D

Cs

(%C

D10

3+ cel

ls o

fC

D45

+ /CD

19– /M

HC

II+ cel

ls)

HI LO0

20

40

60

80

Isle

t pla

smac

ytoi

d de

ndrit

ic c

ells

(%B

220+ /C

D11

c– of

CD

45+ /C

D19

– /MH

CII+ c

ells

)

HGF

Fig. 5. Low accessibility to Ag for CTLs leads to low proliferation of effectors. Intravital observations in the pancreas of RIP-GPxCX3CR1+/GFP mice from the HI and LO scenarios revealed less interactions between islet-specific P14 CTLs and GFP+ APCs in the HI scenario compared with the LO scenario. (A and B) Still frames from represent-ative recordings for HI (A) (movie S4) and for LO (B) (movie S5). (C) Interactions are quantified as the arrest coefficient (fraction of time spent in a nonmoving state). Data are representative of groups of six mice per treatment. (D and E) APC occupancy by islet-specific CTLs and non–islet-specific CTLs was further analyzed in paraformaldehyde- fixed, frozen pancreas sections from RIP-GPxCX3CR1+/GFP mice receiving HI (D) or LO (E) amounts of non–islet-specific CD8+ T cells (n = 4 mice per group). Bars next to images display fractional occupancy of APCs by the indicated cell type. (F to H) The difference in APC occupancy could not be explained by changes in islet APC subsets, as shown in (F) for macrophages, in (G) for migratory DCs, and in (H) for plasmacytoid DCs (n = 3 to 4 mice per group). (I and J) The low level of APC interactions in the HI scenario likely led to a lower level of proliferation of effectors, as assessed by transfer of CellTrace Violet–stained Ly5.1+ P14 CD8+ T cells. n = 5 mice per group. Results are representative of two independent experiments. Scale bars, 10 m. Data are means ± SEM. *P < 0.05, two-tailed unpaired Mann-Whitney U tests.

by guest on March 26, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 9: AUTOIMMUNITY Copyright © 2018 Suppression of diabetes by ...€¦ · Christoffersson et al., Sci. Immunol. 3, eaam6533 2018 23 March 2018 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of

Christoffersson et al., Sci. Immunol. 3, eaam6533 (2018) 23 March 2018

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 12

DISCUSSIONIn this study, we describe a mechanism of immune regulation by CD8+ T cells by which effectors not recognizing local tissue Ags were re-cruited to a site of inflammation, and their abundance at these sites limited tissue damage elicited by CD8+ T cells specific for islet or viral Ags. Non–islet-specific CTLs were found to efficiently, and with ana-tomic specificity, localize to the inflamed islets of Langerhans in models of T1D. When present in high amounts, these cells limited the autoaggressive response from the islet-specific effectors through nonspecific hindrance at the target tissue, resulting in reduced ef-fector T cell proliferation, activation, and down-regulation of effec-tor functions and, ultimately, reduced tissue destruction. We believe that the current study exposes an intrinsic local immune-regulatory mechanism that limits excessive immune responses. As judged by findings in autoimmune and other inflammatory lesions, the CD8+ T cell repertoires at these sites are heterogeneous with only a small fraction of them bearing TCR specificity to auto-Ags or viral epitopes.

T1D is characterized by immune infiltration of the pancreatic islets, resulting in progressive cell decay and eventually onset of hyperglycemia (24). CD8+ T cells constitute the principal immune cell subset within insulitic lesions and have the potential to directly recognize and kill cells (25, 26). Regardless of the molecular events that trigger T cell activation or convey cytotoxic functionality, it has been questioned whether TCR specificity against islet-related epi-topes is a requirement for islet recruitment. Savinov and colleagues (1) showed that preexistent inflammation in the pancreas is dis-pensable for the migration of activated insulin-specific CD8+ T cells across the endothelium. Moreover, endothelial cells were found to actively cross-present islet Ags in the pancreas and thus mediate local T cell adhesion in an Ag-specific manner. In mouse models of allo-transplantation, it was shown that, without preexisting inflamma-tion, only islet-specific activated CTLs or memory CD8+ T cells were able to gain access to tissue through presentation of Ag by endothelial cells or intravascular APC dendrites (27). However, when inflamma-tion was established by the islet-specific cells, non–islet-specific CD8+ T cells gained access through the leukocyte recruitment cascade in-volving signaling through chemokine receptors and integrins (27). In the current study, we show that islet auto-Ags are not required for non–islet-specific T cell accumulation but that local inflamma-tion and T cell activation are critical to this response.

In the RIP-GP mouse model of autoimmune diabetes, only 1 to 2% of the CD8+ T cells in the islet infiltrate were specific to the GP Ag driving the disease (4). This abundance of activated “bystander” CD8+ T cells not recognizing islet Ags has led to hypotheses regard-ing their detrimental effect on cells through the nonspecific release of cytokines without TCR engagement (5, 6). The current study does not argue against the possibility of bystander CTLs promoting cell damage. Instead, we found here that the collective behavior of large groups of activated CTLs not receiving local TCR engagement does not have a disease-accelerating effect. The non–islet-specific CTLs did not differ largely in their expression of surface markers or effector functions dependent on whether they had been transferred to mice in high or low amounts. Neither did they express any evidence of converting into a specialized Treg type. Islet-specific CTLs were the only lymphocyte subset that displayed major differences after being cotransferred with high or low amounts of non–islet-specific CTLs. At the time point when hyperglycemia normally occurs in these models, the amounts of islet-specific CTLs in the pancreas were lower in mice receiving high amounts of non–islet-specific CD8+ T cells. They also

displayed signs of less activation (low expression of CD69), less ef-fector function (low expression of IFN- and high expression of IL-10), and nonresponsiveness (high expression of PD-1 and low expression of KLRG1). All these features thus resulted in the sparing of cells and sustained normoglycemia in these animals.

We examined the cytokine environment in the pancreas, hypoth-esizing that the large influx of “uninvited” non–islet-specific cells would deplete the area of cytokines and metabolic factors, resulting in the impaired proliferation and function of effectors. This was not the case at the organ level because none of the 10 cytokines assessed was dif-ferentially expressed between the two scenarios.

Local interactions within the islets are crucial for the development of effector CD8+ T cells in T1D (28). Interactions between CTLs and APCs in islets during the onset of T1D have also been shown to in-crease effector functions and the production of IFN- (29). We used our imaging model to interrogate whether interference in these con-tacts could explain the decreased effector function when high amounts of non–islet-specific CD8+ T cells were present. Normally in the RIP- GP and RIP-mOVA mouse models, CTL-APC contacts are common in the insulitic lesion (29). In mice receiving high numbers of non–islet-specific CD8+ T cells, these contacts were fewer in vivo, and islet- associated APCs were occupied by a vast majority of transferred non–islet-specific TCR-tg cells or endogenous CD8+ T cells and not by islet-specific effectors. A “scanning” behavior of CTLs interacting with APCs has been described before, and this behavior was unrelated to Ag specificity or Ag presented by either part of the pair (30).

These findings may have relevance for several aspects of human disease. In human T1D, insulitis does not seem to occur as aggres-sively as in current mouse models. Along with other autoimmune dis-orders, T1D has a seemingly slow onset with the appearance of islet autoantibodies (and insulitis?) several years before clinical manifes-tation of the disease. In pancreata from autoantibody-positive organ donors without T1D, evidence for insulitis was found in only 10% of islets or less (31). This has led to hypotheses regarding an appar-ent relapsing-remitting pattern of the disease (32). A low-grade auto-immune response could therefore be interrupted by mechanisms similar to the one presented here by the expansion of large sets of antiviral CTLs to interfere at the insulitic lesion. Certain viral infec-tions have been shown to induce pancreatic inflammation and be pro-tective in mouse models of T1D (33, 34). Nonspecific inflammation through treatment with polyinosinic/polycytidylic acid [poly(I:C)] protects diabetes-prone biobreeding rats and nonobese diabetic mice from disease (35, 36). A recent study has also identified poly(I:C) to condition CD4+ T cells to express FoxP3 and induce a nonspecific bystander suppressor population in tumor and diabetes models (37). In a recent study, the depletion or functional impairment (CCR7- knockout, unable to reach lymph nodes) of Tregs in a mouse model of colitis led to the local induction of IL-10–producing CD8+ T cells that partly controlled inflammation (38).

A recent study found that T cell exhaustion plays an important role in the relapsing-remitting pattern of autoimmune pathologies where the “exhausted appearance” of the nonresponsive CD8+ T cells (PD-1hi, IFN-lo, and IL-10hi) was found to be associated with poor clearance of chronic viral infections but conversely predicted better prognosis in multiple autoimmune diseases (39). Diversifying the CD8+ T cell repertoire at an autoinflammatory site and thereby limiting the access to costimulation for the target-specific effectors may be an intrinsic way for the immune system to self-regulate. This was un-derlined in the current study with the intracerebral LCMV infection

by guest on March 26, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 10: AUTOIMMUNITY Copyright © 2018 Suppression of diabetes by ...€¦ · Christoffersson et al., Sci. Immunol. 3, eaam6533 2018 23 March 2018 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of

Christoffersson et al., Sci. Immunol. 3, eaam6533 (2018) 23 March 2018

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 12

A

CB

ED

F

CD8 OT-I Nuclei CD8 OT-I NucleiPolyclonalOT-I

PolyclonalOT-I

0 5 10 150

20

40

60

80

100

Time post infection (days)

Perc

ent s

urvi

val

Memory pool controlNo cell transfer

1 × 106

1 × 106

1 × 106

1 × 103

1 × 103 OT-I CD8+ T cells

OT-I CD8+ T cells

OT-I CD8+ OT-I CD8+ T cellsT cells

OT-I CD8+ T cells OT-I CD8+ T cells

*

0

20

40

60

80

Cel

ls p

er fi

eld

of v

iew

PolyclonalOT-I

*

*

1 × 103

Fig. 6. Non–viral Ag–specific CTLs dampen inflammation in a mouse model of viral meningitis. (A) Mice were inoculated intracerebrally with LCMV. One group (black line) had been infected with LCMV intraperitoneally 1 month before intracerebral infection and mounted a memory response, and mice were thereby protected. Mice receiving in-tracerebral LCMV without previous induced memory succumbed to the infection 7.25 days (median) after inoculation (red line). In cell transfer experiments similar to the HI and LO scenarios in the T1D models, 1 × 103 or 1 × 106 non–LCMV-specific OT-I CD8+ T cells were transferred and activated in vivo through peptide immunization. High amounts of OT-I led to a significant delay (P = 0.0006, 1 × 103 OT-I versus 1 × 106 OT-I, log-rank test) in time to death (n = 6 mice per group; data are representative of two independent experiments). Brains from the mice were examined 7 days after intracerebral infection. (B and C) No morphological differences or major differences in immune infiltrates could be detected between the groups receiving 1 × 106 (B) or 1 × 103 (C) OT-I CD8+ T cells (n = 4 mice per group). Scale bars, 1 mm. (D and E) Micrographs of periventricular areas in (D) and (E) rep-

resent the squares in (B) and (C) and have been stained using anti-CD8 mAb (gray), Hoechst for nuclei (blue), and OT-I cells expressing GFP (green). Scale bars, 20 m. Bars next to the micrographs represent the clonal distribution of CD8+ T cells. (F) Overall numbers of CD8+ T cell clones per field of view in periventricular regions of brains from mice receiving transfers of 1 × 106 (HI) or 1 × 103 (LO) OT-I CD8+ T cells. *P < 0.05, two-way ANOVA.

by guest on March 26, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 11: AUTOIMMUNITY Copyright © 2018 Suppression of diabetes by ...€¦ · Christoffersson et al., Sci. Immunol. 3, eaam6533 2018 23 March 2018 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of

Christoffersson et al., Sci. Immunol. 3, eaam6533 (2018) 23 March 2018

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

11 of 12

model, where an otherwise aggressive course of disease was slowed by the entrance into the central nervous system by high numbers of a non–LCMV- specific CTL clone. The cause of death in this model is cerebral edema caused by the influx of myelomonocytic cells, which in turn is guided to the site by the secretion of chemokines by the CTLs at the site (40). These data are in line with what was seen in the T1D models in this study, in which islet-specific CTLs shifted their cytokine expression profile in the presence of high numbers of non–islet-specific CTLs. This may be part of the explanation as to why auto-immune attacks are seemingly interrupted and may also be a possible avenue for immune intervention by which nonspecific activation of the immune system could possibly dampen autoaggressive responses.

The antigenic situation in human autoimmunity is complex, with several auto-Ags directing the demise of the target tissue. In this study, we have used autoimmune models in which a single known Ag was driv-ing the disease. Although we have used models with different driver Ags, these situations are still simplified. However, it is evident in hu-man disease and experimental models that T cells lacking specificity to the target tissue will infiltrate inflamed regions (3, 4, 41). Com-bined with our data from a viral infection model, it seems plausible that similar events could also occur in human disease and partly ex-plain viral interference and relapsing-remitting patterns in autoim-munity (34, 39).

In conclusion, this study presents a previously unknown mecha-nism of immune regulation in autoimmunity. The accumulation of effector CD8+ T cells not recognizing local tissue Ags at the pancre-atic islets during the onset of T1D halted an aggressive immune re-sponse, and the animals went into remission with very little evidence of tissue damage. This may reflect inherent mechanisms in the immune system for controlling excessive inflammation and tissue damage and a possible avenue to explore for immune interventions in disease.

MATERIALS AND METHODSAnimalsC57Bl/6J mice were purchased from the Jackson Laboratory. All other strains were bred in-house under specific pathogen–free conditions at the La Jolla Institute for Allergy and Immunology and kept on C57Bl/6J background. Transgenic RIP-LCMV-GP (Berlin strain) (42), RIP- mOVA (43), and RIP-OVAlow (44) mice have been described previ-ously. TCR transgenic strains P14 (45) and OT-I (46) mice were bred on DsRed-, GFP-, or Ly5.1 (B6.SJL-Ptprca Pepcb/BoyJ)–expressing back-grounds. B6.129P-Cx3cr1tm1Litt/J mice (CX3CR1GFP/GFP) were bred with RIP-LCMV-GP mice. Female and male mice were used at 8 to 16 weeks of age. All animal experiments were approved by the Insti-tutional Animal Care and Use Committee at the La Jolla Institute for Allergy and Immunology.

Adoptive cell transfersSingle-cell suspensions were prepared from the spleen and lymph nodes. Red blood cell lysis was performed, and CD8+ T cells were isolated using negative selection (Miltenyi Biotec). Isolated cells were transferred in single-cell suspensions into mice through retro-orbital injections.

Induction of autoimmune diabetesDiabetes was induced in transgenic animals either by infection with 1 × 104 plaque-forming units of recombinant LCMV expressing OVA (LCMV-OVA) or by peptide immunization protocols: Twenty- four hours

after the adoptive cell transfer, 1 mg of GP33–41 (KAVYNFATC, GenScript) and 100 g of OVA257–264 (SIINFEKL, InvivoGen) in 50 g of CpG adjuvant (Integrated DNA Technologies) were injected sub- cutaneously at the same site in the scapular area. The treatment was repeated 48 hours later. On the sixth day after the adoptive cell trans-fer, 500 g of poly(I:C) (InvivoGen) was injected intraperitoneally (fig. S1). Blood glucose levels were measured using test strips (OneTouch Ultra, LifeScan). Mice with blood glucose levels above 250 mg/dl in two consecutive measurements were considered diabetic.

Intravital imagingA previously described protocol for confocal and multiphoton micros-copy of the mouse pancreas (47) was adapted to also incorporate the use of a custom-made immobilization device using vacuum [Fig. 1A and described in (11)]. Briefly, mice were anesthetized with an initial dose of ketamine hydrochloride (90 mg/kg) and xylazine hydrochlo-ride (15 mg/kg), which was iterated as needed. A longitudinal inci-sion was made in the left flank, and the vacuum device was lowered in place to immobilize the pancreas for imaging using a Leica HC Fluotar L 25×/0.95-W objective on a Leica SP5 confocal microscope. The pancreatic islets were in most experiments visualized using the reflected light from the 633-nm laser line.

Flow cytometryAll flow cytometry experiments were run on BD LSR II or Fortessa (BD Biosciences) instruments. Dead cells were discriminated using an Aqua Live/Dead staining kit (Life Technologies). All flow cytome-try data were initially gated on lymphocytes>singlets in two steps>live cells>. Data were analyzed using FlowJo software (Tree Star).

StatisticsData are means ± SEM. Comparisons between the different groups were performed with two-tailed unpaired nonparametric Mann- Whitney U tests (two groups), two-way analysis of variance (ANOVA), or log-rank tests (specified in the respective figure legends). A P value of less than 0.05 was considered significant.

SUPPLEMENTARY MATERIALSimmunology.sciencemag.org/cgi/content/full/3/21/eaam6533/DC1MethodsFig. S1. Ag-driven mouse models of T1D.Fig. S2. CTL activation and expansion is a requirement for pancreatic recruitment.Fig. S3. Immune suppression by non–Ag-specific CD8+ T cells was not due to a “transfer bias.”Fig. S4. No signs of differences in the cytokine environment in the pancreas.Movie S1. Activated CTLs infiltrate islets irrespective of the presence of their cognate Ag.Movie S2. No islet infiltration in the absence of cognate Ag and islet inflammation.Movie S3. Differential behavior of islet-specific CTLs and non–islet-specific CTLs at islets.Movie S4. High amounts of CTLs not recognizing local Ag reduces the number of interactions with APCs.Movie S5. High amounts of CTLs not recognizing local Ag reduces the number of interactions with APCs.Reference (48)

REFERENCES AND NOTES 1. A. Y. Savinov, F. S. Wong, A. C. Stonebraker, A. V. Chervonsky, Presentation of antigen by

endothelial cells and chemoattraction are required for homing of insulin-specific CD8+ T cells. J. Exp. Med. 197, 643–656 (2003).

2. S. A. Sarkar, C. E. Lee, F. Victorino, T. T. Nguyen, J. A. Walters, A. Burrack, J. Eberlein, S. K. Hildemann, D. Homann, Expression and regulation of chemokines in murine and human type 1 diabetes. Diabetes 61, 436–446 (2012).

3. K. T. Coppieters, F. Dotta, N. Amirian, P. D. Campbell, T. W. H. Kay, M. A. Atkinson, B. O. Roep, M. G. von Herrath, Demonstration of islet-autoreactive CD8 T cells in insulitic

by guest on March 26, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 12: AUTOIMMUNITY Copyright © 2018 Suppression of diabetes by ...€¦ · Christoffersson et al., Sci. Immunol. 3, eaam6533 2018 23 March 2018 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of

Christoffersson et al., Sci. Immunol. 3, eaam6533 (2018) 23 March 2018

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

12 of 12

lesions from recent onset and long-term type 1 diabetes patients. J. Exp. Med. 209, 51–60 (2012).

4. M. B. A. Oldstone, K. H. Edelmann, D. B. McGavern, J. T. Cruite, M. J. Welch, Molecular anatomy and number of antigen specific CD8 T cells required to cause type 1 diabetes. PLOS Pathog. 8, e1003044 (2012).

5. D. F. Tough, P. Borrow, J. Sprent, Induction of bystander T cell proliferation by viruses and type I interferon in vivo. Science 272, 1947–1950 (1996).

6. M. S. Horwitz, L. M. Bradley, J. Harbertson, T. Krahl, J. Lee, N. Sarvennick, Diabetes induced by Coxsackie virus: Initiation by bystander damage and not molecular mimicry. Nat. Med. 4, 781–785 (1998).

7. R. K. Gershon, K. Kondo, Cell interactions in the induction of tolerance: The role of thymic lymphocytes. Immunology 18, 723–737 (1970).

8. E. M. Shevach, Regulatory T cells in autoimmmunity. Annu. Rev. Immunol. 18, 423–449 (2000). 9. H. Jiang, S. M. Canfield, M. P. Gallagher, H. H. Jiang, Y. Jiang, Z. Zheng, L. Chess,

HLA-E-restricted regulatory CD8+ T cells are involved in development and control of human autoimmune type 1 diabetes. J. Clin. Invest. 120, 3641–3650 (2010).

10. Z. Dai, S. Zhang, Q. Xie, S. Wu, J. Su, S. Li, Y. Xu, X. C. Li, Natural CD8+CD122+ T cells are more potent in suppression of allograft rejection than CD4+CD25+ regulatory T cells. Am. J. Transplant. 14, 39–48 (2014).

11. M. R. Looney, E. E. Thornton, D. Sen, W. J. Lamm, R. W. Glenny, M. F. Krummel, Stabilized imaging of immune surveillance in the mouse lung. Nat. Methods 8, 91–96 (2011).

12. V. P. Badovinac, J. S. Haring, J. T. Harty, Initial T cell receptor transgenic cell precursor frequency dictates critical aspects of the CD8+ T cell response to infection. Immunity 26, 827–841 (2007).

13. M. M. Martinic, C. Huber, K. Coppieters, J. E. Oldham, A. L. Gavin, M. G. von Herrath, Expression level of a pancreatic neo-antigen in beta cells determines degree of diabetes pathogenesis. J. Autoimmun. 35, 404–413 (2010).

14. M. Rifa’i, Y. Kawamoto, I. Nakashima, H. Suzuki, Essential roles of CD8+CD122+ regulatory T cells in the maintenance of T cell homeostasis. J. Exp. Med. 200, 1123 (2004).

15. S. F. Ziegler, F. Ramsdell, M. R. Alderson, The activation antigen CD69. Stem Cells 12, 456–465 (1994).

16. E. J. Wherry, S. J. Ha, S. M. Kaech, W. N. Haining, S. Sarkar, V. Kalia, S. Subramaniam, J. N. Blattman, D. L. Barber, R. Ahmed, Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 27, 670–684 (2007).

17. J. E. Klementowicz, A. E. Mahne, A. Spence, V. Nguyen, A. T. Satpathy, K. M. Murphy, Q. Tang, Cutting edge: Origins, recruitment, and regulation of CD11c+ cells in inflamed islets of autoimmune diabetes mice. J. Immunol. 199, 27–32 (2017).

18. J. Diana, Y. Simoni, L. Furio, L. Beaudoin, B. Agerberth, F. Barrat, A. Lehuen, Crosstalk between neutrophils, B-1a cells and plasmacytoid dendritic cells initiates autoimmune diabetes. Nat. Med. 19, 65–73 (2013).

19. S. S. Kang, J. Herz, J. V. Kim, D. Nayak, P. Stewart-Hutchinson, M. L. Dustin, D. B. McGavern, Migration of cytotoxic lymphocytes in cell cycle permits local MHC I-dependent control of division at sites of viral infection. J. Exp. Med. 208, 747–759 (2011).

20. B. Calderon, J. A. Carrero, M. J. Miller, E. R. Unanue, Cellular and molecular events in the localization of diabetogenic T cells to islets of Langerhans. Proc. Natl. Acad. Sci. U.S.A. 108, 1561–1566 (2011).

21. K. L. Graham, J. Herz, J. V. Kim, D. Nayak, P. Stewart-Hutchinson, M. L. Dustin, D. B. McGavern, Intra-islet proliferation of cytotoxic T lymphocytes contributes to insulitis progression. Eur. J. Immunol. 42, 1717–1722 (2012).

22. R. M. Sutherland, S. L. Londrigan, J. L. Brady, E. M. Carrington, J. M. Marchingo, S. Heinzel, P. D. Hodgkin, K. L. Graham, T. W. Kay, Y. Zhan, A. M. Lew, Cognate antigen engagement on parenchymal cells stimulates CD8+ T cell proliferation in situ. Nat. Commun. 8, 14809 (2017).

23. C. M. Matullo, K. J. O’Regan, H. Hensley, M. Curtis, G. F. Rall, Lymphocytic choriomeningitis virus-induced mortality in mice is triggered by edema and brain herniation. J. Virol. 84, 312–320 (2010).

24. T. L. van Belle, K. T. Coppieters, M. G. von Herrath, Type 1 diabetes: Etiology, immunology, and therapeutic strategies. Physiol. Rev. 91, 79–118 (2011).

25. K. T. Coppieters, M. G. von Herrath, Histopathology of type 1 diabetes: Old paradigms and new insights. Rev. Diabet. Stud. 6, 85–96 (2009).

26. A. Skowera, R. J. Ellis, R. Varela-Calviño, S. Arif, G. C. Huang, C. Van-Krinks, A. Zaremba, C. Rackham, J. S. Allen, T. I. Tree, M. Zhao, C. M. Dayan, A. K. Sewell, W. W. Unger, J. W. Drijfhout, F. Ossendorp, B. O. Roep, M. Peakman, CTLs are targeted to kill beta cells in patients with type 1 diabetes through recognition of a glucose-regulated preproinsulin epitope. J. Clin. Invest. 118, 3390–3402 (2008).

27. J. M. Walch, Q. Zeng, Q. Li, M. H. Oberbarnscheidt, R. A. Hoffman, A. L. Williams, D. M. Rothstein, W. D. Shlomchik, J. V. Kim, G. Camirand, F. G. Lakkis, Cognate antigen directs CD8+ T cell migration to vascularized transplants. J. Clin. Invest. 123, 2663–2671 (2013).

28. J. Chee, H. J. Ko, A. Skowera, G. Jhala, T. Catterall, K. L. Graham, R. M. Sutherland, H. E. Thomas, A. M. Lew, M. Peakman, T. W. Kay, B. Krishnamurthy, Effector-memory T cells develop in islets and report islet pathology in type 1 diabetes. J. Immunol. 192, 572–580 (2014).

29. R. S. Friedman, R. S. Lindsay, J. K. Lilly, V. Nguyen, C. M. Sorensen, J. Jacobelli, M. F. Krummel, An evolving autoimmune microenvironment regulates the quality of effector T cell restimulation and function. Proc. Natl. Acad. Sci. U.S.A. 111, 9223–9228 (2014).

30. R. M. Kedl, W. A. Rees, D. A. Hildeman, B. Schaefer, T. Mitchell, J. Kappler, P. Marrack, T cells compete for access to antigen-bearing antigen-presenting cells. J. Exp. Med. 192, 1105–1113 (2000).

31. P. In’t Veld, D. Lievens, J. De Grijse, Z. Ling, B. Van der Auwera, M. Pipeleers-Marichal, F. Gorus, D. Pipeleers, Screening for insulitis in adult autoantibody-positive organ donors. Diabetes 56, 2400–2404 (2007).

32. M. von Herrath, S. Sanda, K. Herold, Type 1 diabetes as a relapsing–remitting disease? Nat. Rev. Immunol. 7, 988–994 (2007).

33. C. M. Filippi, E. A. Estes, J. E. Oldham, M. G. von Herrath, Immunoregulatory mechanisms triggered by viral infections protect from type 1 diabetes in mice. J. Clin. Invest. 119, 1515–1523 (2009).

34. T. Boettler, M. von Herrath, Protection against or triggering of Type 1 diabetes? Different roles for viral infections. Expert Rev. Clin. Immunol. 7, 45–53 (2011).

35. D. V. Serreze, K. Hamaguchi, E. H. Leiter, Immunostimulation circumvents diabetes in NOD/Lt mice. J. Autoimmun. 2, 759–776 (1989).

36. D. O. Sobel, D. Goyal, B. Ahvazi, J. W. Yoon, Y. H. Chung, A. Bagg, D. M. Harlan, Low dose poly I:C prevents diabetes in the diabetes prone BB rat. J. Autoimmun. 11, 343–352 (1998).

37. L. J. Thompson, J. F. Lai, A. C. Valladao, T. D. Thelen, Z. L. Urry, S. F. Ziegler, Conditioning of naive CD4+ T cells for enhanced peripheral Foxp3 induction by nonspecific bystander inflammation. Nat. Immunol. 17, 297–303 (2016).

38. Y. I. Kim, B. R. Lee, J. H. Cheon, B. E. Kwon, M. N. Kweon, H. J. Ko, S. Y. Chang, Compensatory roles of CD8+ T cells and plasmacytoid dendritic cells in gut immune regulation for reduced function of CD4+ Tregs. Oncotarget 7, 10947–10961 (2016).

39. E. F. McKinney, J. C. Lee, D. R. W. Jayne, P. A. Lyons, K. G. C. Smith, T-cell exhaustion, co-stimulation and clinical outcome in autoimmunity and infection. Nature 523, 612–616 (2015).

40. J. V. Kim, S. S. Kang, M. L. Dustin, D. B. McGavern, Myelomonocytic cell recruitment causes fatal CNS vascular injury during acute viral meningitis. Nature 457, 191–195 (2009).

41. J. M. Schenkel, K. A. Fraser, V. Vezys, D. Masopust, Sensing and alarm function of resident memory CD8+ T cells. Nat. Immunol. 14, 509–513 (2013).

42. P. S. Ohashi, S. Oehen, K. Buerki, H. Pircher, C. T. Ohashi, B. Odermatt, B. Malissen, R. M. Zinkernagel, H. Hengartner, Ablation of “tolerance” and induction of diabetes by virus infection in viral antigen transgenic mice. Cell 65, 305–317 (1991).

43. C. Kurts, W. R. Heath, F. R. Carbone, J. Allison, J. F. Miller, H. Kosaka, Constitutive class I-restricted exogenous presentation of self antigens in vivo. J. Exp. Med. 184, 923–930 (1996).

44. E. Blanas, F. R. Carbone, J. Allison, J. F. A. P. Miller, W. R. Heath, Induction of autoimmune diabetes by oral administration of autoantigen. Science 274, 1707–1709 (1996).

45. H. Pircher, K. Bürki, R. Lang, H. Hengartner, R. M. Zinkernagel, Tolerance induction in double specific T-cell receptor transgenic mice varies with antigen. Nature 342, 559–561 (1989).

46. K. A. Hogquist, S. C. Jameson, W. R. Heath, J. L. Howard, M. J. Bevan, F. R. Carbone, T cell receptor antagonist peptides induce positive selection. Cell 76, 17–27 (1994).

47. K. Coppieters, N. Amirian, M. von Herrath, Intravital imaging of CTLs killing islet cells in diabetic mice. J. Clin. Invest. 122, 119–131 (2012).

48. M. G. von Herrath, M. Yokoyama, J. Dockter, M. B. Oldstone, J. L. Whitton, CD4-deficient mice have reduced levels of memory cytotoxic T lymphocytes after immunization and show diminished resistance to subsequent virus challenge. J. Virol. 70, 1072–1079 (1996).

Acknowledgments: We are grateful to G. Aguila (La Jolla Institute for Allergy and Immunology) for expert mouse colony management and P. Colby (La Jolla Institute for Allergy and Immunology) for administrative assistance. Funding: G. Christoffersson was supported by a postdoctoral fellowship from the Swedish Research Council. This study was funded by the NIH. Author contributions: G. Christoffersson, G. Chodaczek, K.C., and M.G.v.H. conceptualized the study. G. Christoffersson, G. Chodaczek, S.S.R., and K.C. provided the methodology. G. Christoffersson, G. Chodaczek, S.S.R., and K.C. conducted the investigation. G. Christoffersson performed statistical analysis. G. Christoffersson and M.G.v.H. wrote the original draft of the manuscript. G. Christoffersson, G. Chodaczek, K.C., and M.G.v.H. wrote, reviewed, and edited the manuscript. M.G.v.H. supervised the study. Competing interests: M.G.v.H. and K.C. are employees of Novo Nordisk. All other authors declare that they have no competing interests.

Submitted 23 December 2016Resubmitted 11 September 2017Accepted 30 January 2018Published 23 March 201810.1126/sciimmunol.aam6533

Citation: G. Christoffersson, G. Chodaczek, S. S. Ratliff, K. Coppieters, M. G. von Herrath, Suppression of diabetes by accumulation of non–islet-specific CD8+ effector T cells in pancreatic islets. Sci. Immunol. 3, eaam6533 (2018).

by guest on March 26, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 13: AUTOIMMUNITY Copyright © 2018 Suppression of diabetes by ...€¦ · Christoffersson et al., Sci. Immunol. 3, eaam6533 2018 23 March 2018 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of

pancreatic islets effector T cells in+islet-specific CD8−Suppression of diabetes by accumulation of non

Gustaf Christoffersson, Grzegorz Chodaczek, Sowbarnika S. Ratliff, Ken Coppieters and Matthias G. von Herrath

DOI: 10.1126/sciimmunol.aam6533, eaam6533.3Sci. Immunol. 

model. Together, these results reveal an immune-regulatory role for nonautoreactive CTLs.led to a state of unresponsiveness. A similar form of nonspecific suppression by CTLs was observed in a viral meningitis

islet-specific CTLs in islets reduced the accessibility of islet-specific CTLs to autoantigens, which−The abundance of non accumulation of these ''bystander'' CTLs is associated with decreased activation and proliferation of islet-specific CTLs.

. observed that theet alislet-specific, and their contribution to T1D is not well understood. Christoffersson −non (CTLs) in pancreatic islets, which leads to islet destruction and loss of insulin production. Most of the CTLs in islets are

T cells +Type 1 diabetes (T1D) is associated with the infiltration of islet-specific autoreactive cytotoxic CD8Infiltration inhibition

ARTICLE TOOLS http://immunology.sciencemag.org/content/3/21/eaam6533

MATERIALSSUPPLEMENTARY http://immunology.sciencemag.org/content/suppl/2018/03/22/3.21.eaam6533.DC1

REFERENCES

http://immunology.sciencemag.org/content/3/21/eaam6533#BIBLThis article cites 48 articles, 16 of which you can access for free

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 © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of

by guest on March 26, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from