Notch regulates IL-10 production by T helper 1 cells · T helper 1 (Th1) cells mediate powerful...

6
Notch regulates IL-10 production by T helper 1 cells Sascha Rutz, Marko Janke, Nadine Kassner, Thordis Hohnstein, Manuela Krueger, and Alexander Scheffold* Deutsches Rheuma-Forschungszentrum Berlin, Charite ´ platz 1, 10117 Berlin, Germany Communicated by Klaus Rajewsky, Harvard Medical School, Boston, MA, December 23, 2007 (received for review July 2, 2007) T helper 1 (Th1) cells mediate powerful cellular immune responses. However, if unbalanced, Th1 immunity eventually may cause pathology. Recently, it has been shown that IL-10, an antiinflam- matory cytokine strongly antagonizing Th1-mediated effects, can be produced by Th1 cells and is indeed essential for self-regulation of Th1 immunity. Here, we show that Notch induces IL-10 produc- tion in newly developing and already established Th1 cells via a signal transducer and activator of transcription 4 (STAT4)-depen- dent process. Notch signaling in the presence of the cytokines IL-12 or IL-27 induces Th1 cells to produce large amounts of IL-10 without diminishing IFN- production. Notch-modified Th1 cells completely lose their inflammatory capacity and instead are able to actively suppress a Th1 cell-induced delayed-type hypersensitivity (DTH) reaction in an IL-10-dependent fashion. IL-10 production can be elicited by active forms of all four mammalian Notch receptors but was found to be specific for the Delta-like family of Notch ligands. Dendritic cells (DC) selectively acquire Delta-like 4 expression upon stimulation with various Toll-like receptor (TLR) ligands and con- comitantly induce IL-10 production by Th1 cells in vitro and in vivo. This effect can be selectively reversed by pharmacological inhibi- tors of Notch signaling (-secretase inhibitor). Our data suggest that Notch regulates IL-10 production in Th1 cells by a STAT4- dependent process that converts proinflammatory Th1 cells into T cells with regulatory activity. This pathway may provide unique opportunities for therapeutic intervention in Th1-driven immune diseases and for Th1-associated vaccination strategies. immunoregulation inflammation Notch signaling T helper differentiation T regulatory 1 cells T helper 1 (Th1) cells, characterized by the production of large amounts of IFN-, control inflammatory immune responses and mediate the defense against intracellular pathogens. To prevent pathology associated with uncontrolled activation, the immune system also employs various negative regulatory circuits. One such strategy is the release of immunosuppressive cytokines. First of all, IL-10 has the capacity to potently suppress Th1 effector functions, mainly by preventing the activation of antigen-presenting cells (APCs) (1). Among CD4 T cells, IL-10 originally has been described as a Th2-associated cytokine that antagonizes Th1 re- sponses (1, 2). Subsequently, IL-10 has been shown to be produced also by specialized subsets of regulatory T cells (Treg), such as CD25 FoxP3 Treg (3, 4), and T regulatory (Tr) 1 (5) cells, which are thought to be essential for an effective negative regulation of immune responses. Interestingly, Th1 cells themselves were found to secrete IL-10 (6, 7). Thus, Th1-derived IL-10 recently has been shown to be critical for the prevention of immune pathology in the course of infection with the intracellular pathogens Toxoplasma gondii and Leishmania major (8, 9). These results revealed a self-limiting property of Th1 cells to contain immune reactions independently of regulatory T cells. The molecular pathways leading to IL-10 expression in Th1 cells are only beginning to be understood. In Th2 cells, IL-10 is induced by the main Th2-polarizing cytokine IL-4 by the transcription factor GATA-3 as part of the Th2 differentiation program (10, 11). Repeated stimulation under suboptimal conditions induces IL-10, producing T cells with regulatory capacity. However, these cells express no or reduced levels of IFN- compared with Th1 cells (5, 12, 13). In contrast, Th1 cells generated in vitro contain only a few IL-10-secreting cells. The frequency of these cells can be increased by repeated stimulation in the presence of IL-12 (11, 14, 15). Recently, another member of the IL-12 family of cytokines, IL-27, has been described to induce IL-10 production both in naı ¨ve and effector T cells (16–18). The Notch pathway previously has been implicated in the regu- lation of peripheral T cell activation (19 –21) and the differentiation into various effector T cell populations (22). However, the com- plexity of the Notch system, which is a direct consequence of the numerous potential receptor–ligand interactions of four Notch receptors expressed on T cells and five Notch ligands [Jagged-1, Jagged-2, Delta-like (Dll)-1, Dll-2, and Dll-4] expressed on APCs, so far has prevented the establishment of a clear-cut picture of the role of Notch in peripheral T cell differentiation. Recent data convincingly show that Notch signaling is required for Th2 differ- entiation in vivo (23–27). However, the proposed involvement of Notch in Th1 (23, 28, 29) or likewise regulatory T cell development (30, 31) is less clear. These partially contradictory results strongly suggest that the outcome of Notch-directed T cell differentiation depends on additional, as-yet-uncharacterized, costimulatory sig- nals or certain environmental niches. Here, we identify Notch as a potent inducer of IL-10 both in developing and in already established IFN--secreting Th1 cells. This effect requires costimulation by the cytokines IL-12 or IL-27 that depends on signal transducer and activator of transcription 4 (STAT4) signaling. Our data suggest that Notch regulates IL-10 production by Th1 cells under inf lammatory conditions and thereby facilitates the self-limitation of Th1 immune responses. Results Notch Influences Th Cell Differentiation. To determine the inf luence of T cell-polarizing cytokines on Notch-induced T cell differenti- ation, we stimulated naı ¨ve CD4 T cells under Th1, Th2, or nonpolarizing conditions and retrovirally transduced the cells with constitutively active Notch (N3IC). At day 5 of culture, the trans- duced T cells were isolated according to their GFP expression, restimulated with phorbol 12-myristate 13-acetate (PMA)/ ionomycin, and analyzed for cytokine production. Notch did not grossly alter the cytokine profile under nonpolar- izing or Th2 conditions. As reported earlier (23, 32), IL-4 produc- tion was slightly increased by Notch. No changes were observed in IL-2, IL-5, IL-13, IL-17, or IFN- expression. Notch slightly induced IL-10 under nonpolarizing conditions and further increased the already high levels of IL-10 in Th2 cells. Similarly, under Th1 conditions, Notch only minimally affected expression of the Th1 cytokines IL-2 and IFN-. The most striking observation, however, was a strong IL-10 production by Notch-transduced Th1 cells (Fig. 1A), whereas IL-10 was virtually absent in control Th1 cultures. Author contributions: S.R. and A.S. designed research; S.R., M.J., N.K., T.H., and M.K. performed research; S.R. analyzed data; and S.R. and A.S. wrote the paper. The authors declare no conflict of interest. *To whom correspondence should be addressed. E-mail: [email protected]. This article contains supporting information online at www.pnas.org/cgi/content/full/ 0712102105/DC1. © 2008 by The National Academy of Sciences of the USA www.pnas.orgcgidoi10.1073pnas.0712102105 PNAS March 4, 2008 vol. 105 no. 9 3497–3502 IMMUNOLOGY Downloaded by guest on November 13, 2020

Transcript of Notch regulates IL-10 production by T helper 1 cells · T helper 1 (Th1) cells mediate powerful...

Page 1: Notch regulates IL-10 production by T helper 1 cells · T helper 1 (Th1) cells mediate powerful cellular immune responses. However, if unbalanced, Th1 immunity eventually may cause

Notch regulates IL-10 production by T helper 1 cellsSascha Rutz, Marko Janke, Nadine Kassner, Thordis Hohnstein, Manuela Krueger, and Alexander Scheffold*

Deutsches Rheuma-Forschungszentrum Berlin, Chariteplatz 1, 10117 Berlin, Germany

Communicated by Klaus Rajewsky, Harvard Medical School, Boston, MA, December 23, 2007 (received for review July 2, 2007)

T helper 1 (Th1) cells mediate powerful cellular immune responses.However, if unbalanced, Th1 immunity eventually may causepathology. Recently, it has been shown that IL-10, an antiinflam-matory cytokine strongly antagonizing Th1-mediated effects, canbe produced by Th1 cells and is indeed essential for self-regulationof Th1 immunity. Here, we show that Notch induces IL-10 produc-tion in newly developing and already established Th1 cells via asignal transducer and activator of transcription 4 (STAT4)-depen-dent process. Notch signaling in the presence of the cytokines IL-12or IL-27 induces Th1 cells to produce large amounts of IL-10 withoutdiminishing IFN-� production. Notch-modified Th1 cells completelylose their inflammatory capacity and instead are able to activelysuppress a Th1 cell-induced delayed-type hypersensitivity (DTH)reaction in an IL-10-dependent fashion. IL-10 production can beelicited by active forms of all four mammalian Notch receptors butwas found to be specific for the Delta-like family of Notch ligands.Dendritic cells (DC) selectively acquire Delta-like 4 expression uponstimulation with various Toll-like receptor (TLR) ligands and con-comitantly induce IL-10 production by Th1 cells in vitro and in vivo.This effect can be selectively reversed by pharmacological inhibi-tors of Notch signaling (�-secretase inhibitor). Our data suggestthat Notch regulates IL-10 production in Th1 cells by a STAT4-dependent process that converts proinflammatory Th1 cells into Tcells with regulatory activity. This pathway may provide uniqueopportunities for therapeutic intervention in Th1-driven immunediseases and for Th1-associated vaccination strategies.

immunoregulation � inflammation � Notch signaling � T helperdifferentiation � T regulatory 1 cells

T helper 1 (Th1) cells, characterized by the production of largeamounts of IFN-�, control inflammatory immune responses

and mediate the defense against intracellular pathogens. To preventpathology associated with uncontrolled activation, the immunesystem also employs various negative regulatory circuits. One suchstrategy is the release of immunosuppressive cytokines. First of all,IL-10 has the capacity to potently suppress Th1 effector functions,mainly by preventing the activation of antigen-presenting cells(APCs) (1). Among CD4� T cells, IL-10 originally has beendescribed as a Th2-associated cytokine that antagonizes Th1 re-sponses (1, 2). Subsequently, IL-10 has been shown to be producedalso by specialized subsets of regulatory T cells (Treg), such asCD25�FoxP3� Treg (3, 4), and T regulatory (Tr) 1 (5) cells, whichare thought to be essential for an effective negative regulation ofimmune responses.

Interestingly, Th1 cells themselves were found to secrete IL-10 (6,7). Thus, Th1-derived IL-10 recently has been shown to be criticalfor the prevention of immune pathology in the course of infectionwith the intracellular pathogens Toxoplasma gondii and Leishmaniamajor (8, 9). These results revealed a self-limiting property of Th1cells to contain immune reactions independently of regulatory Tcells.

The molecular pathways leading to IL-10 expression in Th1 cellsare only beginning to be understood. In Th2 cells, IL-10 is inducedby the main Th2-polarizing cytokine IL-4 by the transcription factorGATA-3 as part of the Th2 differentiation program (10, 11).Repeated stimulation under suboptimal conditions induces IL-10,producing T cells with regulatory capacity. However, these cellsexpress no or reduced levels of IFN-� compared with Th1 cells (5,

12, 13). In contrast, Th1 cells generated in vitro contain only a fewIL-10-secreting cells. The frequency of these cells can be increasedby repeated stimulation in the presence of IL-12 (11, 14, 15).Recently, another member of the IL-12 family of cytokines, IL-27,has been described to induce IL-10 production both in naıve andeffector T cells (16–18).

The Notch pathway previously has been implicated in the regu-lation of peripheral T cell activation (19–21) and the differentiationinto various effector T cell populations (22). However, the com-plexity of the Notch system, which is a direct consequence of thenumerous potential receptor–ligand interactions of four Notchreceptors expressed on T cells and five Notch ligands [Jagged-1,Jagged-2, Delta-like (Dll)-1, Dll-2, and Dll-4] expressed on APCs,so far has prevented the establishment of a clear-cut picture of therole of Notch in peripheral T cell differentiation. Recent dataconvincingly show that Notch signaling is required for Th2 differ-entiation in vivo (23–27). However, the proposed involvement ofNotch in Th1 (23, 28, 29) or likewise regulatory T cell development(30, 31) is less clear. These partially contradictory results stronglysuggest that the outcome of Notch-directed T cell differentiationdepends on additional, as-yet-uncharacterized, costimulatory sig-nals or certain environmental niches.

Here, we identify Notch as a potent inducer of IL-10 both indeveloping and in already established IFN-�-secreting Th1 cells.This effect requires costimulation by the cytokines IL-12 or IL-27that depends on signal transducer and activator of transcription 4(STAT4) signaling. Our data suggest that Notch regulates IL-10production by Th1 cells under inflammatory conditions and therebyfacilitates the self-limitation of Th1 immune responses.

ResultsNotch Influences Th Cell Differentiation. To determine the influenceof T cell-polarizing cytokines on Notch-induced T cell differenti-ation, we stimulated naıve CD4� T cells under Th1, Th2, ornonpolarizing conditions and retrovirally transduced the cells withconstitutively active Notch (N3IC). At day 5 of culture, the trans-duced T cells were isolated according to their GFP expression,restimulated with phorbol 12-myristate 13-acetate (PMA)/ionomycin, and analyzed for cytokine production.

Notch did not grossly alter the cytokine profile under nonpolar-izing or Th2 conditions. As reported earlier (23, 32), IL-4 produc-tion was slightly increased by Notch. No changes were observed inIL-2, IL-5, IL-13, IL-17, or IFN-� expression. Notch slightly inducedIL-10 under nonpolarizing conditions and further increased thealready high levels of IL-10 in Th2 cells. Similarly, under Th1conditions, Notch only minimally affected expression of the Th1cytokines IL-2 and IFN-�. The most striking observation, however,was a strong IL-10 production by Notch-transduced Th1 cells (Fig.1A), whereas IL-10 was virtually absent in control Th1 cultures.

Author contributions: S.R. and A.S. designed research; S.R., M.J., N.K., T.H., and M.K.performed research; S.R. analyzed data; and S.R. and A.S. wrote the paper.

The authors declare no conflict of interest.

*To whom correspondence should be addressed. E-mail: [email protected].

This article contains supporting information online at www.pnas.org/cgi/content/full/0712102105/DC1.

© 2008 by The National Academy of Sciences of the USA

www.pnas.org�cgi�doi�10.1073�pnas.0712102105 PNAS � March 4, 2008 � vol. 105 � no. 9 � 3497–3502

IMM

UN

OLO

GY

Dow

nloa

ded

by g

uest

on

Nov

embe

r 13

, 202

0

Page 2: Notch regulates IL-10 production by T helper 1 cells · T helper 1 (Th1) cells mediate powerful cellular immune responses. However, if unbalanced, Th1 immunity eventually may cause

Notch Induces IL-10 in Newly Developing and Established Th1 Cells. Toconfirm the de novo expression of IL-10 in Th1 cells, we performedintracellular cytokine staining (Fig. 1B). Strikingly, Notch inducedIL-10 in �40% of the transduced Th1 cells, whereas �1% ofcontrol cells produced IL-10. Importantly, IL-10 expression wasrestricted to GFP-positive, i.e., Notch-transduced T cells, indicatinga cell-intrinsic effect (Fig. 1B). The frequency of IFN-�-expressingcells was up-regulated by Notch from �50% to 95%. Moreover,intracellular cytokine staining unveiled a strong correlation be-tween IFN-� and IL-10. Indeed, all IL-10-producing T cells exhib-ited an IFN-�/IL-10 double-positive phenotype (Fig. 1C). Similarly,IL-10 mRNA was up-regulated only in Notch-transduced T cells(Fig. 1D). Active forms of all of the Notch receptors (Notch-1 to

Notch-4) were capable of up-regulating IL-10 expression (Fig. 1E).The effect could be reversed by a specific inhibitor of the �-secre-tase complex (GSI) in cells overexpressing a membrane-boundactive Notch (Fig. 1F, NICmem) but not in cells transduced with acytosolic active Notch (NICcyt) that is no longer sensitive to theinhibitor. These data show that the Notch-mediated induction ofIL-10 expression under Th1-polarizing conditions involves theclassical �-secretase-dependent Notch pathway.

To test whether IL-10 also can be induced in already establishedTh1 cells, we Th1-polarized naıve T cells for 1 week and purifiedIFN-�-producing cells by IFN-� capture assay. Then, these cellswere transduced with Notch and cultured for additional 5 daysunder Th1 conditions. As shown in Fig. 2, all cells still expressed

0

2000

4000

6000

8000

10000

12000

14000

Cyt

okin

ee x

p re s

sion

[ pg/

ml]

IL-2IL-4IL-5IL-10IL-13IL-17IFN-γ

Notch control Cyt

okin

ee x

p re s

sion

[pg/

ml ]

Cy t

o kin

ee x

p re s

sion

[pg/

ml]

Notch control Notch control

IL-2IL-4IL-5IL-10IL-13IL-17IFN-γ

IL-2IL-4IL-5IL-10IL-13IL-17IFN-γ

I L-1

0

GFP

IFN

I L-2

I L-4

No t

c h- 3

I C

T 1H

34,9% 47,5% 77,7% 97% 0% 0% 0,9% 0,7%

29,9% 94,5% 72% 65,3% 0% 0% 1,6% 39,2%

A

B

IL-1

0ex

p res

s ing

cel ls

[%]

0

10

20

30

N-1 N-2 N-3 N-4 C

10

20

0

0 GSI5 nM25 nM125 nM

cyt memNotch Notch control

rela

tive

I L-1

0ex

pres

sio n

0

5

10

15

+ - + - N N C CIL-10

Not

ch-3

ICc o

ntro

l

IL- 1

0e x

pres

sing

cells

[%]

T 2Hnon polarized

C

E

IFN

-γ0

2000

4000

6000

8000

10000

0

5000

10000

15000

20000

43,8% 22,1%

0,2%

D

F

Fig. 1. Notch induces IL-10 in developing Th1 cells. (A) Cytokine profile after PMA/ionomycin restimulation of naıve CD4�CD25�CD62L� T cells from C57BL/6mice that had been cultured for 5 days under nonpolarizing, Th1-polarizing, or Th2-polarizing conditions and retrovirally transduced with constitutively activeNotch or GFP alone (control). (B) Intracellular cytokine staining of Notch- or control-transduced T cells cultured under Th1-polarizing conditions for 5 days andPMA/ionomycin restimulation. Percentages indicate frequency of cytokine-producing cells among GFP-positive or GFP-negative cells (gated on CD4� T cells). (C)Intracellular staining for IFN-� and IL-10 on PMA/ionomycin-restimulated T cells (gated on CD4� T cells). (D) IL-10 mRNA levels after PMA/ionomycin restimulationof Notch (N)- or control (C)-transduced T cells sorted for GFP expression. (E) IL-10 expression analyzed by intracellular staining after retroviral transduction withNotch-1 to Notch-4 (N-1 to N-4) of naıve T cells cultured under Th1 conditions and PMA/ionomycin restimulation. C, control transfected. (F) Frequency ofIL-10-producing cells analyzed by intracellular staining after PMA/ionomycin restimulation of naıve T cells from BALB/c mice that had been cultured underTh1-polarizing conditions and retrovirally transduced with a cytosolic (NICcyt) or a membrane-bound (NICmem) form of an active Notch or GFP alone. GSI had beenadded during culture as indicated. (Data are representative of at least three independent experiments each.)

3498 � www.pnas.org�cgi�doi�10.1073�pnas.0712102105 Rutz et al.

Dow

nloa

ded

by g

uest

on

Nov

embe

r 13

, 202

0

Page 3: Notch regulates IL-10 production by T helper 1 cells · T helper 1 (Th1) cells mediate powerful cellular immune responses. However, if unbalanced, Th1 immunity eventually may cause

IFN-�. However, although only 5% of the cells coexpressed IL-10in the control sample, �20% of the Notch-transduced Th1 cellsproduced IL-10. This difference was even more pronounced whenthe secreted cytokines were measured in the culture supernatants.

STAT4 Activation by IL-12 or IL-27 Is Critical for IL-10 Induction byNotch. Next, we determined which additional factors are requiredfor the Notch-mediated IL-10 induction. To exclude a role forfurther costimulatory signals provided by APCs, highly purified Tcells were stimulated in the presence of IL-12 by using plate-boundanti-CD3/anti-CD28. Under these conditions, we still observedcomparable levels of IL-10 in Notch-transduced T cells (Fig. 3A).In contrast, IL-10 production was abrogated in STAT4- but not inIFN-�R-deficient cells (Fig. 3B), supporting the idea that directsignaling by STAT4 is required for IL-10 induction.

Expression of the Th1-specific transcription factor T-bet wasdown-regulated in Notch-transduced cells at day 5 of the culture(Fig. 3C), indicating that T-bet is not critical for IL-10 up-regulation. In accordance with this observation, IL-10 still wasobserved in T-bet-deficient cells although at a slightly reduced level(Fig. 3D). The expression of the Th2 transcription factor GATA3was slightly increased by Notch (Fig. 3C). However, no Th2cytokines, such as IL-4, IL-5, or IL-13, were expressed in Notch-transduced cells under Th1-polarizing conditions (Fig. 1 A and B).FoxP3 was not up-regulated by Notch (data not shown).

IL-27 recently has been described as inducing IL-10 in T cells(16–18). Indeed, when IL-27 was added to naıve T cell cultures, weobserved an expression of IL-10, which was comparable to thatinduced by IL-12 alone (Fig. 3E). However, upon transduction withNotch, the amount of IL-10 was drastically increased. Again, theup-regulation of IL-10 was completely blocked in T cells fromSTAT4 knockout mice. Thus, our data indicate that STAT4 acti-

vation by IL-12 or IL-27 is sufficient for Notch-mediated up-regulation of IL-10 production in Th1 cells.

Notch-Modified Th1 Cells Lose Proinflammatory Capacity and BecomeSuppressive in Vivo in an IL-10-Dependent Manner. To investigate theproinflammatory or antiinflammatory capacity of IL-10-producingNotch-modified Th1 cells in vivo, Notch- or control-transducedovalbumin-specific T cell receptor (OVA-TCR) transgenic Th1cells were adoptively transferred into BALB/c recipient mice. After24 h, a delayed-type hypersensitivity (DTH) reaction was inducedby footpad injection of OVA peptide.

Control-transduced Th1 cells elicited a strong footpad swellingthat peaked at day 1 and then slowly declined but remained clearlydetectable until day 14. In sharp contrast, after a comparable acuteresponse on day 1, swelling rapidly declined in mice that hadreceived Notch-transduced Th1 cells and had ceased completelywithin 72 h (Fig. 4A). These results indicate that Notch-modifiedTh1 cells have lost their potential to maintain inflammatory re-sponses for prolonged periods of time.

To test whether Notch-modified Th1 cells become activelysuppressive, we cotransferred Notch- or control-transduced OVA-specific Th1 cells together with the same number of conventionalOVA-specific Th1 effector cells (Th1eff). Th1 effector-inducedfootpad swelling was further increased by cotransfer of control-transduced Th1 cells, probably because of the increased number ofproinflammatory cells. In contrast, the presence of Notch-transduced Th1 cells significantly reduced footpad swelling, evenwhen compared with Th1 cells transferred alone (Fig. 4B). Thissuppression was almost abrogated by a neutralizing anti-IL10antibody (Fig. 4C).

These data show that Notch induces a functional switch in Th1cells from a proinflammatory into an antiinflammatory phenotype

IFN

IFN

- γ

Control

IL-10

IL-1

0pr

oduc

t ion

[pg/

ml]

N C

5,3%94,7%21,2%78,8%

0

2000

4000

6000

8000

10000

no stain

Notch-3IC

Fig. 2. Notch induces the immunosuppressive cytokine IL-10 in established IFN-�-secreting Th1 cells. IFN-� producers that had been isolated from 1-week-oldTh1 cells were subjected to an additional week of Th1 culture and retroviral transduction with Notch (N) or GFP alone as a control (C). After PMA/ionomycinrestimulation, IFN-� and IL-10 production was analyzed by intracellular staining and in culture supernatants. (Data are representative of two independentexperiments.)

Wildtype STAT4 -/- IFN-γR -/-

0

10

20

30

40

50

Notch Contr.IL-1

0ex

pres

sing

cells

[%]

I L-1

0ex

pres

sio n

[pg/

ml ] 4000

3000

2000

1000

4000

3000

2000

1000

4000

3000

2000

1000

Notch Contr. Notch Contr. Notch Contr.

APC-freeA B

IL-1

0ex

pres

sion

[pg/

ml] 4000

3000

2000

1000

4000

3000

2000

1000

WildtypeIL-27

STAT4 -/-IL-27

rela

tive

mR

NA

e xpr

essi

o n

2,0

1,5

1,0

0,5

N C

GATA-3

500

1000

1500

0,9

0,6

0,3

N C

T-bet

500

1000

1500

I L-1

0e x

pres

sion

[pg/

ml ]

Wildtype T-bet -/-

Notch Contr. Notch Contr. Notch Contr. Notch Contr.

C D E

Fig. 3. Notch-mediated IL-10 inductionstrictly depends on STAT4 activation. (A)Analysis of IL-10 production by intracellularstaining after PMA/ionomycin restimula-tion of naıve T cells transduced with activeNotch or GFP alone as control and culturedfor 5 days cultured under Th1 conditionsand plate-bound anti-CD3/anti-CD28 stim-ulation. (B) IL-10 levels in culture superna-tants of PMA/ionomycin-restimulated iso-lated Notch- or control-transduced T cellsfrom wild-type, STAT4�/�, or IFN-�R�/�

mice that had been cultured for 5 daysunder Th1 conditions. (C) Expression ofGATA-3 and T-bet in isolated Notch (N)- orcontrol (C)-transduced T cells after 5 days ofculture under Th1 conditions. (D and E)IL-10 levels in culture supernatants of PMA/ionomycin-restimulated isolated Notch- orcontrol-transduced T cells from wild-type and T-bet�/� mice that had been cultured for 5 days under Th1 conditions (D) or from wild-type and STAT4�/� micethat had been cultured for 5 days in the presence of IL-27 (E). (Data are representative of at least two independent experiments.)

Rutz et al. PNAS � March 4, 2008 � vol. 105 � no. 9 � 3499

IMM

UN

OLO

GY

Dow

nloa

ded

by g

uest

on

Nov

embe

r 13

, 202

0

Page 4: Notch regulates IL-10 production by T helper 1 cells · T helper 1 (Th1) cells mediate powerful cellular immune responses. However, if unbalanced, Th1 immunity eventually may cause

with the potential to actively suppress inflammatory reactions viaan IL-10-dependent mechanism.

Expression of Notch Receptors and Notch Ligands on T Cells andDendritic Cells (DCs). We then wanted to identify Notch receptorsand Notch ligands that could play a role during physiologic Tcell–APC interactions. To this end, we analyzed the endogenousexpression pattern of these molecules on the mRNA level. OnlyNotch-1 and Notch-2, but not Notch-3 or Notch-4, were expressedby naıve and activated T cells [supporting information (SI) Fig. 7A].

Ex vivo-isolated splenic and lymph node CD11c� DCs expressedlow levels of Jagged-1, and Dll-1 and Dll-4 were not detectable (SIFig. 7B). Because it had been described that Toll-like receptor(TLR) activation can induce Notch ligand expression (23, 33), wealso analyzed the effects of various TLR ligands. Strikingly, LPS(TLR2/4), Flagellin (TLR5), or CpG (TLR9) stimulation stronglyinduced Dll-4 on DCs, whereas the expression of Dll-1 and Jagged-1was largely unaffected (SI Fig. 7B).

CpG Induces Notch-Dependent IL-10 Production in Th1 Cells in Vitroand in Vivo. To test whether the expression of Dll-4 by activated DCsmay have functional relevance, we cocultured naıve OVA-TCRtransgenic T cells with purified DCs in the presence of CpG, IL-12,or both. After 5 days, IFN-� and IL-10 production was analyzed byintracellular staining after PMA/ionomycin restimulation. Withoutfurther additions, only �1% of the T cells expressed IL-10. CpG orIL-12 alone increased the frequency of IL-10-producing cells to 5%and 8%, respectively. After the combined addition of IL-12 andCpG, �14% of the T cells produced IL-10, which was strictlycoexpressed with IFN-�. Importantly, under all three cultureconditions, GSI clearly reduced IL-10 production by 50–80%,whereas the expression of IFN-� remained largely unaffected(Fig. 5A).

Next, we determined whether the Notch-mediated IL-10 induc-tion also can be observed in vivo. To this end, we transferred naıveOVA-TCR transgenic T cells into BALB/c recipients and immu-nized them with OVA peptide/CpG in the absence or presence ofGSI. On day 5, the coexpression of IFN-� and IL-10 was investi-gated by intracellular staining (Fig. 5B). After CpG/OVA admin-istration, �15% of the OVA-specific T cells produced IFN-�.Approximately 30% of these cells coexpressed IL-10. Strikingly, theproduction of IL-10 was completely blocked in mice that hadreceived GSI, whereas IFN-� production was enhanced. These datasuggest that Notch signaling triggered by Dll-4 expressed on acti-vated DCs can induce IL-10 production by Th1 cells in a physio-logical immune response.

Notch Ligands of the Dll Family Mediate IL-10 Induction. To confirmthe importance of Dll-4 for Notch-mediated IL-10 up-regulationduring T cell–APC interactions, we generated A20 B cell linesoverexpressing the Notch ligands Jagged-1, Dll-1, and Dll-4 (Fig.6A). Naıve OVA-TCR transgenic T cells were cultured with theligand-expressing A20 cells for 1 week under Th1-polarizing con-ditions. Approximately 6% and 8% of the T cells expressed IL-10after coculture with Dll-1- and Dll-4-transduced A20 cells, respec-tively, whereas Jagged-1- and GFP-transduced A20 cells did notinduce significant IL-10 production. In accordance with publisheddata (23), Jagged-1-expressing A20 cells reduced IFN-� productionby �20% compared with control A20 cells, whereas Dll-1 and Dll-4slightly increased the frequency of IFN-�-positive cells. Both IL-10induction by Dll-1 or Dll-4 and suppression of IFN-� by Jagged-1were completely reversed by GSI (Fig. 6B). After a second week ofcoculture under Th1 conditions, Dll-4 or Dll-1 expressing A20

**

time [d]

foot

pad

swel

li ng

[ mm

]

time [d]

foot

pad

swel

ling

[mm

]

0 1 2 3 4 5 6 7 8 9 10

0.00

0.25

0.50

0.75

1.00

1.25

TH1(Notch)+TH1eff

TH1(control)+TH1eff

TH1eff**

**** ****

*

*

TH1(Notch)TH1(control)

0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16

0.00

0.25

0.50

0.75

1.00

1.25

**** ** ** **

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

0 1 2 3 4 5 6 7 8 9 10

0.00

0.25

0.50

0.75

1.00

1.25

1.50

1.75

foot

p ad

swel

li ng

[mm

]

time [d]

TH1(control)+TH1eff + IsotypeTH1(control)+TH1eff + anti-IL-10TH1(Notch)+TH1eff + IsotypeTH1(Notch)+TH1eff + anti-IL-10

* * ***

A B C

Fig. 4. Notch-modified Th1 cells lose their proinflammatory capacity and become suppressive in a DTH response. OVA-TCR transgenic Notch- or control-transduced Th1 cells were adoptively transferred into BALB/c mice. After 24 h, a DTH reaction was induced by footpad injection of OVA peptide (filled symbols).Feet of control mice were injected with PBS (open symbols). (A and B) Notch- or control-transduced Th1 cells were transferred either alone (A) or cotransferredwith conventional Th1 cells (B). (C) Mice received an anti-IL-10 antibody or isotype control before cotransfer of Notch- or control-transduced Th1 cells withconventional Th1 effector cells (Th1eff). Statistical significance was estimated by two-tailed Mann–Whitney test (*, P � 0.05; **, P � 0.01). (Data arerepresentative of two independent experiments, six animals per group.)

-G

SI

CpG

IFN

IL-10

A

B

IL-12 IL-12/CpG

+G

SI

10,94 4,46

1,37

24,07 0,51

0,46OV

A-T

CR

+ GSI- GSI

42,24 1,14

0,01

60,69 7,94

0,06

60,41 2,12

0,02

38,08 0,72

0,01

56,51 13,31

0,00

44,52 4,06

0,05

67,68 6,63

0,06

IFN

- γ

IL-10

CD4

no addition

Fig. 5. CpG induces Notch-dependent IL-10 production in Th1 cells in vitroand in vivo. (A) IL-10 and IFN-� expression analyzed by intracellular staining ofnaıve T cells after 5 days of coculture with CD11c� DCs from spleen and lymphnodes and addition of IL-12, CpG, or IL-12/CpG in the presence or absence ofGSI (gated on CD4� T cells). (Data are representative of at least three inde-pendent experiments.) (B) IL-10 and IFN-� expression analyzed by intracellularstaining after PMA/ionomycin restimulation of OVA-TCR transgenic T cellsthat had been isolated from draining lymph nodes after adoptive transfer ands.c. immunization with OVA/CpG in the presence or absence of GSI. (Data arerepresentative of three independent experiments, three animals per group.)

3500 � www.pnas.org�cgi�doi�10.1073�pnas.0712102105 Rutz et al.

Dow

nloa

ded

by g

uest

on

Nov

embe

r 13

, 202

0

Page 5: Notch regulates IL-10 production by T helper 1 cells · T helper 1 (Th1) cells mediate powerful cellular immune responses. However, if unbalanced, Th1 immunity eventually may cause

induced �24% of IL-10-producing Th1 cells. Again, Jagged-1 wasnot able to elicit IL-10 expression (Fig. 6C). These data show thatNotch signaling triggered by ligands of the Dll family induces IL-10production in Th1 cells, especially on repeated stimulation.

DiscussionOur study identifies a unique role for Notch in effector T celldifferentiation. In addition to its previously described potential todirectly instruct Th1 or Th2 cell differentiation, our data suggestthat Notch also modifies the way T cells respond to the proinflam-matory cytokines IL-12 or IL-27 by selectively enhancing IL-10production and thereby conveying antiinflammatory capacity tootherwise proinflammatory Th1 cells.

So far, Notch has been described to instruct the differentiation ofnaıve T cells into various effector T cell lineages. An essential rolefor Notch in Th2 differentiation has been demonstrated convinc-ingly in vitro and in vivo. Th2 responses are severely impaired in theabsence of Notch (25, 32), and both IL-4 and GATA-3 have beenidentified as being direct transcriptional targets of Notch (23, 24,27). Although we do not see drastic changes in the overall cytokineprofile upon overexpression of constitutively active Notch in naıveT cells, the slight increase in Th2 cytokine expression we observeunder nonpolarizing and under Th2 conditions is in accordancewith these data.

In contrast, the precise function of Notch in Th1 differentiationis more obscure. Notch can induce or enhance Th1 differentiationin vitro (23, 28), and the Th1 transcription factor T-bet has beenidentified as a Notch target gene (29). Although some studies reportNotch to be important for Th1 differentiation in vivo (29, 34), othersfind it to be dispensable (25) or even observe enhanced IFN-� levelsafter abrogation of Notch signaling (32). We found increased IFN-�production upon Notch activation only under Th1-polarizing con-

ditions. In contrast, in vivo inhibition of Notch signaling by GSIresulted in enhanced IFN-� production. Thus, the impact of Notchon Th1 differentiation, as analyzed by the expression of Th1cytokines, varies drastically between different experimental sys-tems. It can only be speculated that it is influenced by furthercostimulatory signals or the cytokine environment.

However, our data demonstrate that the drastic enhancement ofIL-10 expression in IFN-�-producing T cells represents the moststriking effect of Notch on T cell differentiation, which we consis-tently observed under various experimental conditions in vitro andin vivo. The induction of these IFN-�/IL-10 double producers is notrestricted to naıve T cells but is equally effective in alreadyestablished Th1 cells.

Importantly, IL-10 induction by Notch strictly depends on simul-taneous STAT4 activation either by IL-12 or IL-27. Both cytokines,but especially IL-27, recently have been implicated in the regulationof IL-10 expression by Th1 cells (11, 14–18). In agreement withprevious reports, we find basal IL-10 expression in the absence ofNotch signaling when naıve T cells are cultured with either IL-12 orIL-27 alone. However, in either case, IL-10 production is drasticallyenhanced by Notch. This difference is best exemplified by the factthat Notch-modified Th1 cells completely lose their proinflamma-tory capacity and instead suppress Th1-mediated immune reactionsin vivo via an IL-10-dependent process.

The strong synergistic effect of Notch on IL-12/IL-27-mediatedIL-10 induction suggests that it acts as a master regulator thatsensitizes proinflammatory Th1 cells to develop into IFN-��IL-10�

T cells with antiinflammatory potential in response to canonicalinflammatory cytokine signals.

The IL-27-mediated IL-10 induction has been described asdepending on STAT1 and/or STAT3 (17, 18) but to be independentof STAT4 (18). In contrast, the Notch-dependent up-regulation ofIL-10 by IL-27 is abolished in STAT4-deficient T cells, suggestingthat different molecular pathways are triggered when IL-27 actsalone or in combination with Notch. Apart from the STAT4activation, we have no indication for the involvement of other majorTh1-associated factors because Notch induced comparable levels ofIL-10 in T cells from IFN-� receptor-deficient and T-bet-deficientmice.

We demonstrate here that IFN-��IL-10� T cells also are inducedin vivo during immune responses after CpG/OVA vaccination.Importantly, IL-10 but not IFN-� production is completely blockedby GSI, clearly indicating a Notch-dependent process. Our data areconsistent with the previously described occurrence of IFN-��IL-10� T cells in vivo (7, 35, 36). Recently, it has been demonstratedthat, indeed, IL-10 produced by Th1 cells is critical in protecting thehost from Th1-induced pathology (8, 9). It is tempting to speculatethat Notch also may play a role in these situations. In support forthis idea, we and others find that ligands of the Dll family, inparticular Dll4, are strongly induced on DCs by several proinflam-matory microbial stimuli known to drive Th1 immunity (23, 33, 34).The selective induction of Dll-4 on activated DCs in response toCpG stimulation and other microbial stimuli identifies Dll-4 as thepresumptive physiologic ligand triggering Notch-dependent IL-10production. In contrast, Jagged-1 is similarly expressed by restingand activated DCs, indicating a lesser functional importance underthese circumstances. Indeed, we were able to confirm functionaldifferences between Jagged and Dll ligands by their respectiveoverexpression in an APC line. Jagged-1 decreased IFN-� produc-tion in a GSI-sensitive manner as reported earlier (23), whereasDll-1 and Dll-4 elicited IL-10 expression and even enhanced IFN-�in Th1 cells. Overall, our observations support the emerging viewthat Jagged-1 drives Th2 development, whereas Dll ligands aremore important for Th1 cell differentiation (23, 34). However, morerefined experimental systems, such as DC-specific conditionalknockout mice of the various Notch ligands, are required to identifythe partner(s) mediating the critical physiological interaction.

A

+G

SI

GFP Dll-1 Dll-4 Jagged-1

noG

SI

GFP Dll-1 Dll-4 Jagged-1B

C

IL-10

64,48 8,10

0,81

63,07 6,20

0,98

33,52 1,86

1,60

46,58 2,41

1,14

61,31 1,83

0,63

57,32 1,99

0,65

48,33 1,46

0,48

51,84 1,29

0,36IFN

68,88 0,77

0,05

55,42 24,26

0,44

56,52 23,91

0,22

72,27 1,02

0,00

IL-10

IFN

Fig. 6. Notch ligands of the Dll family induce IL-10 production by Th1 cells.(A) Staining for Notch ligands on A20 B cells that had been lentivirallytransduced with Notch ligands Dll-4, Dll-1, Jagged-1, or GFP alone as control.(B and C) Analysis of IL-10 and IFN-� production after PMA/ionomycin restimu-lation of purified naıve OVA-TCR transgenic T cells after coculture with Notchligand-expressing A20 B cells and stimulation with OVA peptide under Th1-polarizing conditions for 1 week (B) or 2 weeks (C). In some samples, GSI hadbeen added (gated on CD4� T cells). (Data are representative of three inde-pendent experiments.)

Rutz et al. PNAS � March 4, 2008 � vol. 105 � no. 9 � 3501

IMM

UN

OLO

GY

Dow

nloa

ded

by g

uest

on

Nov

embe

r 13

, 202

0

Page 6: Notch regulates IL-10 production by T helper 1 cells · T helper 1 (Th1) cells mediate powerful cellular immune responses. However, if unbalanced, Th1 immunity eventually may cause

In summary, our data suggest that the Notch pathway plays animportant role for the autoregulation of Th1 immunity. Notchinstructs T cells to respond to the canonical proinflammatorycytokines IL-12 and IL-27 by expressing the antiinflammatorymediator IL-10, which allows self-limitation of Th1 immune reac-tions. This finding defines a role for Notch as a molecular switchbetween proinflammatory and antiinflammatory Th1 cell function,thus representing a promising target for therapeutic intervention ininflammatory diseases and for improved vaccination strategies.

Materials and MethodsMice. BALB/c, OVA-TCRtg/tg DO11.10, C57BL/6 (BgVV Berlin), T-bet�/� (kind giftfrom J. Penninger, Vienna, Austria), IFN-�R�/� (kind gift from T. Schuler, Berlin,Germany), and STAT4�/� (The Jackson Laboratory) mice were housed underspecific pathogen-free (SPF) conditions and used at 8 to 12 weeks of age.

Cell Purification. Naıve CD4� T cells were isolated by CD4 preenrichment (Au-toMACS, Miltenyi Biotec) and FACS sorting of CD4�CD62L�CD25� T cells (FACSAria, BD Biosciences). CD11c� DCs were isolated via FACS. APCs were sorted viaMACS using MHC class II microbeads. For isolation of IFN-�-producing T cells, theIFN-� secretion assay (Miltenyi Biotec) was used according to the manufacturer’sprotocols and sorted by FACS.

T Cell Stimulation. Naıve T cells were cultured with irradiated MHCII� cells (30 Gy,1:4 ratio), A20 cells (120 Gy, 2:1 ratio), or CD11c� DC (15:1 ratio) as APCs andanti-CD3 (0.5 �g/ml)/CD28 (1 �g/ml) or OVA323–339 peptide (0.2 �g/ml) was added.rIL-12 (10 ng/ml; R&D Systems) and anti-IL4 (10 �g/ml) (for Th1), anti-IL-12/anti-IFN-� (10 �g/ml) and rIL-4 (100 ng/ml) (for Th2), anti-IL-4 and rIL-27 (10 ng/ml) (forIL-27) were added. Where indicated, 500 nM of CpG (ODN1826, Invivogen) or 125nM GSI (Insolution GSI X, Calbiochem) was added. For APC-free systems, plate-bound anti-CD3 (5 �g/ml) and soluble anti-CD28 (1 �g/ml) were used.

BioPlex Analysis of Cytokine Production. Concentrations of IL-2, IL-4, IL-5, IL-10,IL-13, IL-17, and IFN-� were measured in culture supernatants with the BioPlexsystem (Bio-Rad Laboratories) according to the manufacturer’s protocol. Fordetection, phycoerythrin-conjugated streptavidin (SA-PE) (Invitrogen) was used.

Immunization. BALB/c mice were transferred with 1 � 107 CD62L� OVA-TCRtransgenic T cells (DO11.10) and immunized s.c. with 50 �g of OVA peptide and20 �g of CpG (Invivogen). GSI DBZ (synthesized by Syncom) was used at 10 �M/kgin 0.1% Tween 80/0.5% Methocel 60 HG (Sigma-Aldrich) injected daily i.p.

DTH. Th1 cells (5 � 105) were transferred alone or together with the same numberof Notch- or control-transduced cells into naıve BALB/c mice. Neutralizing anti-IL-10 antibody (JES5–2A5–7) was injected i.p. Then, 24 h later, 250 ng of OVA323–

339 peptide in incomplete Freund’s adjuvant (IFA) (Sigma) was injected into theleft footpad, and PBS/IFA was injected into the right footpad. Swelling wasmeasured by an Oditest micrometer gauge in a blinded fashion.

Real-Time Quantitative RT-PCR. Real-time PCR was performed with the LightCy-cler instrument and the FastStart DNA Master SYBR Green I kit (Roche Diagnos-tics). Cycling program (10 min at 95°C followed by 40 cycles of 15 s at 95°C, 15 s at60°C (for Dll-1, IL-10, and FoxP3), 65°C (for UbcH5B, Jagged-1, Dll-4, Notch-1,Notch-3, T-bet, and GATA-3), or 67°C (for Notch-2 and Notch-4), and 15 s at 72°C).For primer sequences, see SI Materials and Methods. Data were evaluated withLightCycler softwareversion3.5.28 (RocheDiagnostics)andthesecondderivativemaximum algorithm.

ACKNOWLEDGMENTS. We thank Katharina Raba and Toralf Kaiser for cellsorting and A. Hamann and M. Loehning for critical reading of the manuscript.S.R. was supported by a grant from the Boehringer Ingelheim Fonds. This workwas supportedbyDeutscheForschungsgemeinschaftGrantsSFB633andSFB650,and by European Community Grant MUGEN LSHG-CT-2005-005203.

1. Moore KW, de Waal MR, Coffman RL, O’Garra A (2001) Interleukin-10 and theinterleukin-10 receptor. Annu Rev Immunol 19:683–765.

2. Abbas AK, Murphy KM, Sher A (1996) Functional diversity of helper T lymphocytes.Nature 383:787–793.

3. Asseman C, Mauze S, Leach MW, Coffman RL, Powrie F (1999) An essential role forinterleukin 10 in the function of regulatory T cells that inhibit intestinal inflammation.J Exp Med 190:995–1004.

4. de la Rosa M, Rutz S, Dorninger H, Scheffold A (2004) Interleukin-2 is essential forCD4�CD25� regulatory T cell function. Eur J Immunol 34:2480–2488.

5. Groux H, et al. (1997) A CD40� T-cell subset inhibits antigen-specific T-cell responsesand prevents colitis. Nature 389:737–742.

6. Assenmacher M, Schmitz J, Radbruch A (1994) Flow cytometric determination ofcytokines in activated murine T helper lymphocytes: Expression of interleukin-10 ininterferon-gamma and in interleukin-4-expressing cells. Eur J Immunol 24:1097–1101.

7. Gerosa F, et al. (1999) CD4(�) T cell clones producing both interferon-gamma andinterleukin-10 predominate in bronchoalveolar lavages of active pulmonary tubercu-losis patients. Clin Immunol 92:224–234.

8. Jankovic D, et al. (2007) Conventional T-bet(�)Foxp3(�) Th1 cells are the major sourceof host-protective regulatory IL-10 during intracellular protozoan infection. J Exp Med204:273–283.

9. Anderson CF, Oukka M, Kuchroo VJ, Sacks D (2007) CD4(�)CD25(�)Foxp3(�) Th1 cellsare the source of IL-10-mediated immune suppression in chronic cutaneous leishman-iasis. J Exp Med 204:285–297.

10. Zhu J, et al. (2004) Conditional deletion of Gata3 shows its essential function in T(H)1-T(H)2 responses. Nat Immunol 5:1157–1165.

11. Chang HD, et al. (2007) Expression of IL-10 in Th memory lymphocytes is conditional onIL-12 or IL-4, unless the IL-10 gene is imprinted by GATA-3. Eur J Immunol 37:807–817.

12. Barrat FJ, et al. (2002) In vitro generation of interleukin 10-producing regulatoryCD4(�) T cells is induced by immunosuppressive drugs and inhibited by T helper type1 (Th1)- and Th2-inducing cytokines. J Exp Med 195:603–616.

13. Jonuleit H, Schmitt E, Schuler G, Knop J, Enk AH (2000) Induction of interleukin10-producing, nonproliferating CD4(�) T cells with regulatory properties by repetitivestimulation with allogeneic immature human dendritic cells. J Exp Med 192:1213–1222.

14. Gerosa F, et al. (1996) Interleukin-12 primes human CD4 and CD8 T cell clones for highproduction of both interferon-gamma and interleukin-10. J Exp Med 183:2559–2569.

15. Meyaard L, Hovenkamp E, Otto SA, Miedema F (1996) IL-12-induced IL-10 productionby human T cells as a negative feedback for IL-12-induced immune responses. J Im-munol 156:2776–2782.

16. Awasthi A, et al. (2007) A dominant function for interleukin 27 in generating inter-leukin 10-producing antiinflammatory T cells. Nat Immunol 8:1380–1389.

17. Fitzgerald DC, et al. (2007) Suppression of autoimmune inflammation of the centralnervous system by interleukin 10 secreted by interleukin 27-stimulated T cells. NatImmunol 8:1372–1379.

18. Stumhofer JS, et al. (2007) Interleukins 27 and 6 induce STAT3-mediated T cell pro-duction of interleukin 10. Nat Immunol 8:1363–1371.

19. Eagar TN, et al. (2004) Notch 1 signaling regulates peripheral T cell activation. Immu-nity 20:407–415.

20. Adler SH, et al. (2003) Notch signaling augments T cell responsiveness by enhancingCD25 expression. J Immunol 171:2896–2903.

21. Rutz S, Mordmuller B, Sakano S, Scheffold A (2005) Notch ligands Delta-like1, Delta-like4 and Jagged1 differentially regulate activation of peripheral T helper cells. EurJ Immunol 35:2443–2451.

22. Osborne BA, Minter LM (2007) Notch signaling during peripheral T-cell activation anddifferentiation. Nat Rev Immunol 7:64–75.

23. Amsen D, Blander JM, Lee GR, Tanigaki K, Honjo T, Flavell RA (2004) Instruction ofdistinct CD4 T helper cell fates by different notch ligands on antigen-presenting cells.Cell 117:515–526.

24. Amsen D, et al. (2007) Direct regulation of gata3 expression determines the T helperdifferentiation potential of notch. Immunity 27:89–99.

25. Tu L, et al. (2005) Notch signaling is an important regulator of type 2 immunity. J ExpMed 202:1037–1042.

26. Tanaka S, et al. (2006) The interleukin-4 enhancer CNS-2 is regulated by Notch signalsand controls initial expression in NKT cells and memory-type CD4 T cells. Immunity24:689–701.

27. Fang TC, Yashiro-Ohtani Y, Del BC, Knoblock DM, Blacklow SC, Pear WS (2007) Notchdirectly regulates Gata3 expression during T helper 2 cell differentiation. Immunity27:100–110.

28. Maekawa Y, et al. (2003) Delta1-Notch3 interactions bias the functional differentiationof activated CD4� T cells. Immunity 19:549–559.

29. Minter LM, et al. (2005) Inhibitors of gamma-secretase block in vivo and in vitro Thelper type 1 polarization by preventing Notch upregulation of Tbx21. Nat Immunol6:680–688.

30. Hoyne GF, et al. (2000) Serrate1-induced notch signaling regulates the decision be-tween immunity and tolerance made by peripheral CD4(�) T cells. Int Immunol12:177–185.

31. Yvon ES, et al. (2003) Over expression of the Notch ligand, Jagged-1 induces alloan-tigen-specific human regulatory T cells. Blood 102:3815–3821.

32. Tanigaki K, et al. (2004) Regulation of alphabeta/gammadelta T cell lineage commit-ment and peripheral T cell responses by Notch/RBP-J signaling. Immunity 20:611–622.

33. Napolitani G, Rinaldi A, Bertoni F, Sallusto F, Lanzavecchia A (2005) Selected Toll-likereceptor agonist combinations synergistically trigger a T helper type 1-polarizingprogram in dendritic cells. Nat Immunol 6:769–776.

34. Skokos D, Nussenzweig MC (2007) CD8� DCs induce IL-12-independent Th1 differen-tiation through Delta 4 Notch-like ligand in response to bacterial LPS. J Exp Med 204:1525–1531.

35. Jankovic D, Kullberg MC, Hieny S, Caspar P, Collazo CM, Sher A (2002) In the absenceof IL-12, CD4(�) T cell responses to intracellular pathogens fail to default to a Th2pattern and are host protective in an IL-10(�/�) setting. Immunity 16:429–439.

36. Shaw MH, et al. (2006) Tyk2 negatively regulates adaptive Th1 immunity by mediatingIL-10 signaling and promoting IFN-gamma-dependent IL-10 reactivation. J Immunol176:7263–7271.

3502 � www.pnas.org�cgi�doi�10.1073�pnas.0712102105 Rutz et al.

Dow

nloa

ded

by g

uest

on

Nov

embe

r 13

, 202

0