+ All Categories
Home > Documents > NOD2 is a negative regulator of Toll-like receptor 2–mediated T … · 2005. 5. 2. · NOD2 to...

NOD2 is a negative regulator of Toll-like receptor 2–mediated T … · 2005. 5. 2. · NOD2 to...

Date post: 31-Aug-2020
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
9
ARTICLES 800 VOLUME 5 NUMBER 8 AUGUST 2004 NATURE IMMUNOLOGY Mutations in CARD15, which encodes nucleotide-binding oligomer- ization domain 2 (NOD2), underlie the occurrence of intestinal inflammatory disease in a substantial subgroup of patients with Crohn disease 1–4 . NOD2 is a member of the NOD–leucine-rich repeat (LRR) protein family (also called the CATERPILLER family), whose members share a tripartite domain structure consisting of a C-terminal peptide recognition (LRR) domain, a central NOD domain and an N terminus made up of protein-protein interaction domains, such as caspase recruitment domains (CARDs) or pyrin domains 5–7 . NOD2 is expressed intracellularly in antigen-present- ing cells (APCs) 8 and through its C-terminal LRR it allows these cells to recognize and react to a component of bacterial peptidogly- can (PGN), muramyl dipeptide (MDP) 9,10 . This is shown by the fact that when NOD2-transfected human embryonic kidney (HEK) 293 cells are exposed to MDP, NOD2 enters into CARD-CARD inter- actions with the serine-threonine kinase RICK, resulting in activa- tion of the transcription factor NF-κB 11,12 . Thus, the ability of NOD2 to act as an intracellular receptor for MDP suggests that NOD2 functions in activation of the innate immune response to bacterial products. NOD1, a close relative of NOD2, probably has a similar function through the ability of its LRR to recognize another derivative of PGN, γ-D-glutamyl-meso-diaminopimelic acid (iE-DAP) 13,14 . PGN is recognized independently of its MDP or iE-DAP compo- nents by cell surface Toll-like receptor 2 (TLR2), and cells that lack TLR2 are refractory to PGN stimulation and fail to show PGN- induced cytokine secretion 15 . PGN signaling via TLR2, as in the case of MDP signaling via NOD2, results in NF-κB activation. However, this occurs via a distinct pathway common to TLRs NOD2 is a negative regulator of Toll-like receptor 2–mediated T helper type 1 responses Tomohiro Watanabe 1 , Atsushi Kitani 1 , Peter J Murray 2 & Warren Strober 1 The mechanism by which mutations in CARD15, which encodes nucleotide-binding oligomerization domain 2 (NOD2), cause Crohn disease is poorly understood. Because signaling via mutated NOD2 proteins leads to defective activation of the transcription factor NF-κB, one proposal is that mutations cause deficient NF-κB-dependent T helper type 1 (T H 1) responses and increased susceptibility to infection. However, this idea is inconsistent with the increased T H 1 responses characteristic of Crohn disease. Here we used Card15 –/– mice to show that intact NOD2 signaling inhibited Toll-like receptor 2–driven activation of NF- κB, particularly of the NF-κB subunit c-Rel. Moreover, NOD2 deficiency or the presence of a Crohn disease–like Card15 mutation increased Toll-like receptor 2–mediated activation of NF-κB–c-Rel, and T H 1 responses were enhanced. Thus, CARD15 mutations may lead to disease by causing excessive T H 1 responses. 1 Mucosal Immunity Section, Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 10 Room 11N238, 10 Center Drive, Bethesda, Maryland 20892, USA. 2 Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, USA. Correspondence should be addressed to W.S. ([email protected]). Published online 27 June 2004; doi:10.1038/ni1092 involving recruitment of interleukin 1 receptor (IL-1R)–associated kinase to the receptor via the adaptor protein MyD88 (refs. 15,16). Because PGN is recognized by TLR2 and its subcomponents by NOD receptors with both forms of recognition inducing NF-κB activation, TLR2 and NOD signaling could be linked so that the extracellular and intracellular responses to pathogen components are coordinated. The mechanism by which CARD15 mutations cause susceptibility to Crohn disease is poorly understood. In vitro studies using HEK 293 cells transfected with NOD2 molecules bearing Crohn disease–asso- ciated CARD15 mutations demonstrate defective NF-κB activation after stimulation of cells with bacterial ligands 1,9,17,18 . Thus, deficient NF-κB activation in response to bacterial products followed by covert infection with certain bacteria is considered to be one mechanism by which CARD15 mutations could lead to disease. However, this idea is inconsistent with the fact that NF-κB activation is essential in the generation of cytokines such as IL-12 and other T helper type 1 (T H 1) cytokines known to underlie the inflammation of Crohn disease 19–25 . Consequently, we sought an explanation for the association of CARD15 mutations and Crohn disease that better accounts for the observation that Crohn disease is marked by increased, not decreased, NF-κB activation. We used NOD2-deficient (Card15 –/– ) mice to show that NOD2 sig- naling normally inhibits the TLR2-driven T H 1 response by regulating NF-κB signaling. In addition, we show that in the absence of NOD2 signaling, such as that occurring in the presence of a Card15 muta- tion, NOD2-mediated inhibition is abrogated, resulting in increased TLR2-mediated NF-κB activation and more IL-12 production. Thus, the CARD15 mutation leads to increased T H 1 cytokine production. © 2004 Nature Publishing Group http://www.nature.com/natureimmunology
Transcript
Page 1: NOD2 is a negative regulator of Toll-like receptor 2–mediated T … · 2005. 5. 2. · NOD2 to act as an intracellular receptor for MDP suggests that NOD2 functions in activation

A RT I C L E S

800 VOLUME 5 NUMBER 8 AUGUST 2004 NATURE IMMUNOLOGY

Mutations in CARD15, which encodes nucleotide-binding oligomer-ization domain 2 (NOD2), underlie the occurrence of intestinalinflammatory disease in a substantial subgroup of patients withCrohn disease1–4. NOD2 is a member of the NOD–leucine-richrepeat (LRR) protein family (also called the CATERPILLER family),whose members share a tripartite domain structure consisting of aC-terminal peptide recognition (LRR) domain, a central NODdomain and an N terminus made up of protein-protein interactiondomains, such as caspase recruitment domains (CARDs) or pyrindomains5–7. NOD2 is expressed intracellularly in antigen-present-ing cells (APCs)8 and through its C-terminal LRR it allows thesecells to recognize and react to a component of bacterial peptidogly-can (PGN), muramyl dipeptide (MDP)9,10. This is shown by the factthat when NOD2-transfected human embryonic kidney (HEK) 293cells are exposed to MDP, NOD2 enters into CARD-CARD inter-actions with the serine-threonine kinase RICK, resulting in activa-tion of the transcription factor NF-κB11,12. Thus, the ability ofNOD2 to act as an intracellular receptor for MDP suggests thatNOD2 functions in activation of the innate immune response tobacterial products. NOD1, a close relative of NOD2, probably hasa similar function through the ability of its LRR to recognizeanother derivative of PGN, γ-D-glutamyl-meso-diaminopimelicacid (iE-DAP)13,14.

PGN is recognized independently of its MDP or iE-DAP compo-nents by cell surface Toll-like receptor 2 (TLR2), and cells that lackTLR2 are refractory to PGN stimulation and fail to show PGN-induced cytokine secretion15. PGN signaling via TLR2, as in thecase of MDP signaling via NOD2, results in NF-κB activation.However, this occurs via a distinct pathway common to TLRs

NOD2 is a negative regulator of Toll-like receptor 2–mediated T helper type 1 responsesTomohiro Watanabe1, Atsushi Kitani1, Peter J Murray2 & Warren Strober1

The mechanism by which mutations in CARD15, which encodes nucleotide-binding oligomerization domain 2 (NOD2), cause Crohn disease is poorly understood. Because signaling via mutated NOD2 proteins leads to defective activation of thetranscription factor NF-κB, one proposal is that mutations cause deficient NF-κB-dependent T helper type 1 (TH1) responses andincreased susceptibility to infection. However, this idea is inconsistent with the increased TH1 responses characteristic of Crohndisease. Here we used Card15–/– mice to show that intact NOD2 signaling inhibited Toll-like receptor 2–driven activation of NF-κB, particularly of the NF-κB subunit c-Rel. Moreover, NOD2 deficiency or the presence of a Crohn disease–like Card15 mutationincreased Toll-like receptor 2–mediated activation of NF-κB–c-Rel, and TH1 responses were enhanced. Thus, CARD15 mutationsmay lead to disease by causing excessive TH1 responses.

1Mucosal Immunity Section, Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 10 Room11N238, 10 Center Drive, Bethesda, Maryland 20892, USA. 2Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee38105, USA. Correspondence should be addressed to W.S. ([email protected]).

Published online 27 June 2004; doi:10.1038/ni1092

involving recruitment of interleukin 1 receptor (IL-1R)–associatedkinase to the receptor via the adaptor protein MyD88 (refs. 15,16).Because PGN is recognized by TLR2 and its subcomponents byNOD receptors with both forms of recognition inducing NF-κBactivation, TLR2 and NOD signaling could be linked so that theextracellular and intracellular responses to pathogen componentsare coordinated.

The mechanism by which CARD15 mutations cause susceptibilityto Crohn disease is poorly understood. In vitro studies using HEK 293cells transfected with NOD2 molecules bearing Crohn disease–asso-ciated CARD15 mutations demonstrate defective NF-κB activationafter stimulation of cells with bacterial ligands1,9,17,18. Thus, deficientNF-κB activation in response to bacterial products followed by covertinfection with certain bacteria is considered to be one mechanism bywhich CARD15 mutations could lead to disease. However, this idea isinconsistent with the fact that NF-κB activation is essential in thegeneration of cytokines such as IL-12 and other T helper type 1 (TH1)cytokines known to underlie the inflammation of Crohn disease19–25.Consequently, we sought an explanation for the association ofCARD15 mutations and Crohn disease that better accounts for theobservation that Crohn disease is marked by increased, not decreased,NF-κB activation.

We used NOD2-deficient (Card15–/–) mice to show that NOD2 sig-naling normally inhibits the TLR2-driven TH1 response by regulatingNF-κB signaling. In addition, we show that in the absence of NOD2signaling, such as that occurring in the presence of a Card15 muta-tion, NOD2-mediated inhibition is abrogated, resulting in increasedTLR2-mediated NF-κB activation and more IL-12 production. Thus,the CARD15 mutation leads to increased TH1 cytokine production.

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urei

mm

unol

ogy

Page 2: NOD2 is a negative regulator of Toll-like receptor 2–mediated T … · 2005. 5. 2. · NOD2 to act as an intracellular receptor for MDP suggests that NOD2 functions in activation

A RT I C L E S

NATURE IMMUNOLOGY VOLUME 5 NUMBER 8 AUGUST 2004 801

RESULTSPGN-stimulated TH1 cytokine production in Card15–/– miceAPCs can digest the TLR2 ligand bacterial PGN and generate thePGN component MDP, a specific agonist for NOD2 (refs. 9,10). Thissuggests that PGN can stimulate cells through both cell surface TLR2and cytoplasmic NOD2. Given this possibility, we addressed the ques-tion of whether NOD2 signaling affects the production of cytokinesinduced by PGN.

In initial studies, we stimulated total splenocytes purified fromCard15–/– mice26 with various TLR ligands (lipopolysaccharide (LPS), aTLR4 ligand; PGN and Pam3Cys, TLR2 ligands; double-stranded RNA(dsRNA), a TLR3 ligand; loxoribine, a TLR7 ligand; and CpG, a TLR9ligand) or the NOD2 agonist MDP and determined cytokine concentra-tions induced by such stimulation. Production of the p40 chain of IL-12(IL-12p40) and (even more so) the p40-p35 heterodimer of IL-12 (IL-12p70) was greatly enhanced in cultures of PGN-stimulated splenocytesfrom Card15–/– mice compared with splenocytes from wild-type mice(Fig. 1, top). We found no increase when NOD2-deficient cells werestimulated by other TLR ligands, which is consistent with previous dataobtained using Card15–/– bone marrow–derived macrophages26 (Fig. 1,top). In contrast, stimulation with MDP significantly decreased produc-tion of IL-12p40 (P = 0.0230) and tumor necrosis factor (TNF; P =0.0366). In addition, increased PGN-induced IL-12 secretion by NOD2-deficient splenocytes was associated with significant increases in pro-duction of both interferon-γ(IFN-γ; P = 0.0010) and IL-18 (P = 0.0296),but no increase in IL-10 production (Fig. 1, middle and bottom). For

10 µg/ml of PGN, the endotoxin concentration was less than 0.5 pg/ml,as determined by a Limulus amebocyte lysate assay27. Thus, the resultsobtained were unlikely to be a result of LPS contamination. In addition,in the presence of MDP, the pure (synthetic) TLR2 agonists Pam3Cysand Pam3CSK4 produced similar results: increased TH1 cytokine secre-tion in Card15–/– mice (Fig. 2). These studies show that, in contrast toMDP stimulation of NOD2-deficient cells, PGN stimulation enhancedproduction of the TH1 cytokines, IL-12, IFN-γand IL-18.

Splenic subpopulations producing TH1 cytokinesWe next determined the types of cells producing increased TH1cytokines in Card15–/– mice. Initially, we assessed PGN-induced pro-duction of IL-12, IFN-γ and IL-18 by Card15–/– splenocyte subpopu-lations depleted of B cells, T cells or macrophages expressing CD11b.Depletion of T cells or B cells did not reduce the increased PGN-induced production of IL-12 or IL-18, whereas macrophage depletiondid reduce production of IL-12p40, IL-12p70 and IL-18 (Fig. 3a). Incontrast, PGN-induced production of IFN-γ was decreased by deple-tion of T cells or macrophages (Fig. 3a).

To confirm these results, we assessed cytokine production by LPS-,PGN- and MDP-stimulated splenic CD11b+ cells. Only PGN-induced IL-12p40 and IL-12p70 production by CD11b+ cells wasenhanced in cells from Card15–/– mice compared with cells fromwild-type mice (Fig. 3b). Enhancement of IL-12p70 production wasgreater than that of IL-12p40 production, suggesting that PGN stimu-lation has a preferential effect on IL-12p35 production in Card15–/–

CD11b+ cells. Consistent with these findings,PGN-stimulated IL-12p35 mRNA expression,as measured by RT-PCR, was notablyincreased in Card15–/– splenic CD11b+ cells(Fig. 3c). We were unable to detect by RT-PCRanalysis IL-23p19 expression in CD11b+ cellsstimulated with PGN or LPS (data notshown), showing that NOD2 signaling is notinvolved in IL-23 production. In addition,there was no difference in TLR2 expression insplenic CD11b+ cells stimulated with PGN(Fig. 3d), Pam3Cys or Pam3CSK4 (data notshown) for wild-type versus Card15–/– mice.

We next analyzed IL-12Rβ1 and IL-12Rβ2chain expression in CD4+ T cells, becauseexpression of these receptors on CD4+ T cells(especially the β2 receptor) is essential forIFN-γ production. PGN stimulation of totalsplenocytes from Card15–/– mice enhancedIL-12Rβ1 and IL-12Rβ2 expression in CD4+ Tcells compared with expression in wild-typecells (Fig. 3e). In contrast, stimulation withLPS or dsRNA did not lead to preferentialenhancement of IL-12Rβ1 and IL-12Rβ2expression in Card15–/– cells. Stimulation ofsplenic CD4+ T cells by IL-12 (5 ng/ml) andIL-18 (1 ng/ml) did not induce differences inIFN-γ production by CD4+ T cells from wild-type and Card15–/– mice (wild-type, 63.6 ±11.7 ng/ml; Card15–/–, 79.5 ± 12 ng/ml). Thesedata suggest that in the absence of NOD2 sig-naling, stimulation with PGN enhanced IL-12production by CD11b+ cells, which led toincreased IFN-γ production by CD4+ T cellsthrough IL-12Rβ2 chain signaling.

Figure 1 Splenocytes from Card15–/– mice show enhanced IL-12 and IFN-γ production after stimulationwith PGN. Total splenocytes obtained from wild-type and Card15–/– mice were stimulated with LPS (TLR4ligand; 1 µg/ml), PGN (TLR2 ligand; 10 µg/ml), MDP (NOD2 ligand; 10 µg/ml), Pam3Cys (TLR2 ligand;500 ng/ml), dsRNA (TLR3 ligand; 25 µg/ml), loxoribine (TLR7 ligand; 100 µM) or CpG (TLR9 ligand; 1 µM). Culture supernatants were collected at 48 h and analyzed for cytokine production by ELISA; resultsare presented as means ± s.d. *, P < 0.05 and **, P < 0.01, compared with wild-type mice. N.D., notdetected. Control concentrations of cytokines in cells without stimulation: IL-12p40, wild-type mice, 118 ± 3 pg/ml, and Card15–/– mice, 147 ± 10 pg/ml; IFN-γ, wild-type mice, 136 ± 11 pg/ml, andCard15–/– mice, 100 ± 14 pg/ml; TNF, wild-type mice, 32 ± 9 pg/ml, and Card15–/– mice, 49 ± 10 pg/ml;IL-12p70, IL-18 and IL-10, below level of detection.

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urei

mm

unol

ogy

Page 3: NOD2 is a negative regulator of Toll-like receptor 2–mediated T … · 2005. 5. 2. · NOD2 to act as an intracellular receptor for MDP suggests that NOD2 functions in activation

A RT I C L E S

802 VOLUME 5 NUMBER 8 AUGUST 2004 NATURE IMMUNOLOGY

Inhibition of TLR2-mediated IL-12 production by NOD2Increased PGN-induced TH1 cytokine secretion by NOD2-deficientsplenocytes could be due to negative regulation of the PGN responseby MDP-NOD2 signaling. To explore this, we compared the cytokineresponses of wild-type and Card15–/– whole-spleen cell populationsstimulated by chemically pure (synthetic) TLR2 ligands (ligands with-out an MDP subcomponent) alone and in the presence of the NOD2agonist MDP. We found no differences in IL-12p40, IL-12p70 andIFN-γ production by wild-type and Card15–/– cells stimulated withsynthetic TLR2 ligands alone (Fig. 2). In contrast, wild-type cells butnot in Card15–/– cells showed decreased production of these cytokines

when spleen cells were stimulated with both TLR2 ligands and MDP,particularly at a high MDP concentration (Fig. 2). This suggests thatMDP signals via NOD2 to reduce TLR2 signaling

In additional studies, we stimulated purified splenic CD11b+

APCs from wild-type and Card15–/– mice with a panel of TLR lig-ands alone or in the presence of increasing concentrations of MDP.Production of IL-12p40 and, to an even greater extent, IL-12p70by wild-type CD11b+ cells stimulated by the TLR2 ligands PGNand Pam3Cys was significantly decreased in a dose-dependent wayby the addition of MDP (PGN, P = 0.0010; Pam3Cys, P = 0.0307;Fig. 4). Stimulation of these cells by other non-TLR2 ligands was

Figure 3 TH1 cytokine production and TLR2 and IL-12R expression by purified spleen subpopulationsfrom wild-type and Card15–/– mice. (a) Splenocytesamples were obtained from Card15–/– mice andwere left nondepleted or were depleted of B cells, T cells or CD11b cells, then were stimulated withLPS (1 µg/ml), PGN (10 µg/ml) or MDP (10 µg/ml);cytokine production was analyzed as described inFigure 1. *, P < 0.05 and **, P < 0.01, comparedwith nondepleted splenocyte samples. Results are presented as means ± s.d. N.D., not detected.(b) IL-12p40 and IL-12p70 production bypositively selected splenic CD11b+ cells stimulatedfor 48 h with LPS (1 µg/ml), PGN (10 µg/ml) orMDP (10 µg/ml). *, P < 0.05 and ** P < 0.01,compared with cells from wild-type mice. Resultsare presented as means ± s.d. N.D., not detected.Control concentrations of cytokines in cells withoutstimulation: IL-12p40, wild-type mice, 132 ± 14pg/ml, and Card15–/– mice, 128 ± 10 pg/ml; IL-12p70, below the level of detection. (c) RT-PCRanalysis of IL-12p35 (Il12a) and IL-12p40 (Il12b)expression in splenic CD11b+ cells stimulated for 5 h with LPS (1 µg/ml) or PGN (10 µg/ml). (d) TLR2 expression on CD11b+ cells. Purifiedsplenic CD11b+ cells were stimulated with PGN for16 h, stained with fluorochrome-labeled antibodies and then analyzed by flow cytometry. Green, orange and red lines show medium alone, 1 µg/ml of PGN and 10 µg/ml of PGN, respectively. (e) IL-12Rβ1 and IL-12Rβ2 chain expression on splenic CD4+ T cells; total splenocytes were stimulated for 24 h with LPS (1 µg/ml), PGN (10 µg/ml) or dsRNA (25 µg/ml), stained with fluorochrome-labeled antibodies and then analyzed by flow cytometry. Analysis gate was set on CD4+

propidium iodide–negative cells. Green and red lines show wild-type and Card15–/– mice, respectively. Arrows (d,e) indicate increasing fluorescence intensity.

a c

d

e

b

Figure 2 Splenocytes from wild-type mice but not Card15–/– mice show decreased TH1 cytokineproduction after costimulation with TLR2 andNOD2 agonists. Total splenocytes purified fromwild-type and Card15–/– mice were stimulated with pure synthetic TLR2 ligands (Pam3Cys orPam3CSK4; 500 ng/ml) and MDP (0, 10 and 100 µg/ml). Cells were also stimulated with PGN(1 and 10 µg/ml). Culture supernatants werecollected at 48 h and analyzed by ELISA forcytokine production; results are presented asmeans ± s.d. *, P < 0.05 and **, P < 0.01,compared with wild-type mice. N.D., not detected.Control concentrations of cytokines in cells withoutstimulation: IL-12p40, wild-type mice, 147 ± 8pg/ml, and Card15–/– mice, 156 ± 13 pg/ml; IFN-γ,IL-12p70 and IL-18, below the level of detection.

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urei

mm

unol

ogy

Page 4: NOD2 is a negative regulator of Toll-like receptor 2–mediated T … · 2005. 5. 2. · NOD2 to act as an intracellular receptor for MDP suggests that NOD2 functions in activation

A RT I C L E S

NATURE IMMUNOLOGY VOLUME 5 NUMBER 8 AUGUST 2004 803

unaffected by the addition of MDP (Fig. 4). In contrast, the addi-tion of MDP did not change IL-12 production by Card15–/–

CD11b+ cells stimulated by any of the TLR ligands, including theTLR2 ligands. These results provide evidence that MDP stimula-tion via NOD2 has an inhibitory effect on TLR2-induced inductionof IL-12 production as well as on ‘downstream’ TH1 cytokines.Moreover, this inhibitory effect was not found in Card15–/– spleno-cytes, thus explaining the increased TH1 cytokine production bythese splenocytes after stimulation by PGN or synthetic TLR2 lig-ands plus MDP.

PGN-induced systemic IL-12 response in Card15–/– miceThe data reported so far predict that an enhanced PGN-induced TH1cytokine response by NOD2-deficient spleen cells should be mir-rored by a corresponding abnormality in NOD2-deficient mice. Totest this possibility, we determined the IL-12 response of wild-typeand Card15–/– mice after intravenous injection of PGN or a controlTLR stimulant, LPS. Intravenous injection of 30 or 300 µg of LPS didnot induce differences in serum IL-12p40 or IL-12p70 concentra-tions between these mice (Fig. 5). In contrast, intravenous injectionof PGN (at the higher concentration tested, 300 µg/mouse) induceda significant increase in the serum concentration of IL-12 (both p40and p70) at 2 or 4 h after challenge (IL-12p40, P = 0.0157; IL-12p70,P = 0.0038; Fig. 5). Thus, PGN stimulation increased IL-12 produc-tion both in vitro and in vivo.

Activation of NF-κB in Card15–/– APCsAs activation of NF-κB is an intersection point of both TLR- andNOD2-mediated signaling, we determined the extent and pattern ofNF-κB subunit translocation to the nucleus in wild-type andCard15–/– cells. Accordingly, we isolated nuclear proteins from APC-rich splenic adherent cell samples after stimulation with LPS, PGNand MDP and quantified NF-κB subunit activity using a semiquanti-tative method based on the binding of the extract to an NF-κB con-sensus sequence followed by detection of bound component withsubunit-specific antibodies. Binding of all tested NF-κB subunits toconsensus sequences was enhanced with nuclear extracts from PGN-stimulated Card15–/– cells compared with extracts from wild-typecells (Fig. 6a). Such enhancement was especially evident with respectto the NF-κB subunit c-Rel., whose binding was increased by a factorof 3.5 relative to the increase noted with NF-κB subunit p65. In con-trast, there were no differences in the binding of any NF-κB subunitwith extracts from LPS-stimulated wild-type and Card15–/– cells,consistent with previous examination of this pathway in bone mar-row–derived macrophages26.

In complementary studies, we confirmed this finding with elec-trophoretic mobility shift assay (EMSA). LPS or PGN stimulation ofsplenic adherent cells induced multiple NF-κB complexes (C1–C6),and MDP stimulation induced two weak NF-κB complexes (C7 andC8; Fig. 6b). In supershift assays, the addition of antibody to p65 (anti-p65) blocked the formation of C1, C2, C4, C5 and C7 complexes,

a

b

Figure 5 Card15–/– mice show enhanced IL-12 production after systemic administration of PGN. Wild-type and Card15–/– mice were given 30 or 300 µg of LPS orPGN intravenously, after which serum samples were collected (time points, horizontal axes). Serum concentrations of IL-12p40 and IL-12p70 were determinedby ELISA. *, P < 0.05 and **, P < 0.01, compared with cytokine concentrations in wild-type mice differences. Results are presented as means ± s.d.

Figure 4 Wild-type spleen macrophages show dose-dependent suppression of TLR2-induced IL-12 production mediated by MDP costimulation and a lack of effect ofcostimulation on IL-12 production induced by stimulants of other TLRs. Splenic CD11b+

cells from wild-type and Card15–/– mice werestimulated for 48 h with LPS (1 µg/ml), PGN(10 µg/ml), Pam3Cys (500 ng/ml), dsRNA (25 µg/ml), loxoribine (100 µM) or CpG (1 µM)in the presence of varying concentrations ofMDP (key); culture supernatants were thenanalyzed by ELISA for the production of IL-12p40 (a) and IL-12p70 (b).*, P < 0.05 and**, P < 0.01, compared with the concentrationof cytokines in the absence of MDP differences.Results are presented as means ± s.d. Controlconcentrations of cytokines in cells withoutstimulation; IL-12p40, wild-type mice, 107 ± 9pg/ml, and Card15–/– mice, 118 ± 17 pg/ml; IL-12p70, below the level of detection.

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urei

mm

unol

ogy

Page 5: NOD2 is a negative regulator of Toll-like receptor 2–mediated T … · 2005. 5. 2. · NOD2 to act as an intracellular receptor for MDP suggests that NOD2 functions in activation

A RT I C L E S

804 VOLUME 5 NUMBER 8 AUGUST 2004 NATURE IMMUNOLOGY

whereas the addition of anti-c-Rel blocked the formation of C1 and C4complexes, and anti-p50 blocked the formation of C1, C3, C4, C6, C7and C8 complexes. This indicated that C1 and C4 complexes consistedof c-Rel–p50 and p65-p50 heterodimers; C2 and C5 complexes con-sisted of p65 heterodimers; and C3, C6 and C8 complexes consisted ofa p50-p50 homodimer. Although wild-type and Card15–/– cells stimu-lated with LPS showed no substantial difference in the nuclear translo-cation of any of the NF-κB subunits, stimulation of these cells withPGN resulted in considerably enhanced translocation of all NF-κBsubunits in Card15–/– cells compared with wild-type cells (Fig. 6b).Consistent with our previous data, c-Rel translocation was notablyincreased in the Card15–/– cells, as shown by the strong band intensityof c-Rel-specific supershift complex obtained with extracts from thesecells. The increased NF-κB translocation induced by PGN is in con-trast to the NF-κB translocation induced by MDP, which was almostabsent in Card15–/– cells. These data indicate that the absence ofNOD2 signaling is accompanied by bothquantitative and qualitative changes in NF-κBsubunit translocation, particularly by c-Rel,in PGN-stimulated cells.

Activation and translocation of c-Rel isrequired for IL-12 production28,29. We there-fore determined whether selective c-Rel inhi-bition in PGN-stimulated NOD2-deficientAPCs leads to normalization of IL-12 pro-duction. We transfected splenic CD11b+ cellsfrom wild-type and Card15–/– mice with c-Rel-specific small interfering RNA (siRNA)and stimulated the transfected cells with LPSor PGN. Transfection of siRNA substantiallydecreased c-Rel expression in whole-cellextracts from Card15–/– CD11b+ cells (Fig. 7a).Inactivation of c-Rel was associated withdecreased PGN-induced IL-12p40 and IL-12p70 production to concentrations found in mock-transfected wild-type cells (Fig. 7a).Moreover, transfection of c-Rel siRNAblocked IL-12p35 mRNA expression by PGN-stimulated Card15–/– CD11b+ cells (data notshown). In contrast, IL-12p70 productionwas not significantly decreased by transfec-tion of siRNA targeting p65, despite efficient

suppression of this protein (P = 0.244; Fig. 7b). Possible effects ofinterferon responses induced by siRNA in this study are unlikely,because dsRNA stimulation did not alter cytokine production andNF-κB activity in Card15–/– mice26 (Figs. 1 and 4). These studies ofNF-κB translocation suggest that enhanced IL-12 production inPGN-stimulated APCs from Card15–/– mice is linked to preferentialactivation and translocation of c-Rel by these cells.

IL-12 production by repleted Card15–/– macrophagesNext, we determined if the aberrant TH1 signaling in Card15–/– APCsis also found with CARD15 mutations in Crohn disease. To examinethis question, we generated a mouse construct, L980fs, that corre-sponded to the human LRR frameshift CARD15 mutationL1007fsinsC (with frameshift (fs) and insertion (ins) at the indicatedsite). We then transfected Card15–/– CD11b+ cells with a hemaggluti-nating virus of Japan (HVJ) envelope vector containing wild-type

Figure 7 Neutralization of c-Rel by siRNA inhibits TLR2-mediated IL-12 production by splenic CD11b+ cells from Card15–/– mice. Splenic CD11b+ cells were incubated with transfection reagent only(Mock) or were transfected with siRNA specific for c-Rel (a) or p65 (b), followed by stimulation withLPS (1 µg/ml) or PGN (10 µg/ml). Culture supernatants collected at 48 h were analyzed by ELISA for IL-12p40 and IL-12p70 secretion.*, P < 0.05 and **, P < 0.01, compared with the cytokinesproduced by mock-transfected cells. Whole-cell extracts were prepared 6 h after PGN stimulation andimmunoblots used anti-c-Rel, anti-p65 and anti-actin. Graphed results are presented as means ± s.d.

a b

a b

Figure 6 Stimulation of splenic adherent cells with PGN enhances translocation of c-Rel into thenucleus. (a) Splenic adherent cells obtained from wild-type and Card15–/– mice were stimulated for 2 h with LPS (1 µg/ml), PGN (10 µg/ml) or MDP (10 µg/ml), after which nuclear extracts were purifiedand expression of NF-κB subunits in nuclear extracts was determined. **, P < 0.01, compared withcells from wild-type mice. Results are presented as absorbance at 450 nm (A450), means ± s.d. (b) EMSA of nuclear extracts from wild-type and Card15–/– splenic adherent cells stimulated withLPS, PGN or MDP. For supershift analyses, anti-p65, anti-c-Rel and anti-p50 were used. Gray, blackand open arrowheads show supershifted complexes specific to p65, c-Rel and p-50, respectively. Ab, antibody. + or –, presence or absence of reagent.

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urei

mm

unol

ogy

Page 6: NOD2 is a negative regulator of Toll-like receptor 2–mediated T … · 2005. 5. 2. · NOD2 to act as an intracellular receptor for MDP suggests that NOD2 functions in activation

A RT I C L E S

NATURE IMMUNOLOGY VOLUME 5 NUMBER 8 AUGUST 2004 805

Card15 or the mutated L980fs Card15. We confirmed expression ofexogenous wild-type and truncated NOD2 proteins by immunoblotanalysis using cell extracts of Card15–/– CD11b+ cells (Fig. 8a). Theband size of the truncated NOD2 protein was slightly smaller thanthat of wild-type protein. Card15–/– CD11b+ cells transfected withwild-type Card15 showed greatly reduced PGN-induced IL-12p40and IL-12p70 production and increased MDP-induced IL-12p40production (Fig. 8b,c). In contrast, Card15–/– CD11b+ cells trans-fected with L980fs Card15 still showed increased PGN-induced IL-12production, and this could not be reversed by MDP, consistent withprevious results (Fig. 4.). The IL-12p70 concentration was below thelevel of detection in culture of CD11b+ cells stimulated with MDP.Finally, transfection of wild-type or L980fs Card15 into Card15–/–

CD11b+ cells did not change LPS-induced IL-12 production. Thesefindings suggest that a frameshift Card15 mutation, like NOD2 defi-ciency, leads to enhanced TH1 cytokine production.

In further studies we sought to determine whether overexpression ofhuman wild-type CARD15 or mutated (L1007fsinsC) CARD15 wouldhave different effects on TLR2-mediated NF-κB activation in a com-pletely ‘reconstituted’ cell system. For this, we transfected the HT-29intestinal cell line, which does not normally express either NOD2 (ref. 30) or TLR2 (ref. 31), with combinations of the following: a con-struct expressing the promoter for the gene encoding NF-κB linked to aluciferase reporter gene; a construct expressing TLR2; a constructexpressing CARD15 or L1007fs CARD15; and a construct expressing thegene encoding β-galactosidase (for purposes of correcting for transfec-tion efficiency). MDP induced a luciferase signal in cells expressing theNF-κB–luciferase reporter with wild-type CARD15 but not in cellsexpressing the reporter gene with the mutated CARD15 (Fig. 8d). Usingcells expressing the NF-κB–luciferase reporter and TLR2 along witheither wild-type CARD15 or mutated CARD15, we showed that

although PGN induced a luciferase signal in the presence of wild-typeCARD15, it induced a significantly higher luciferase signal in the pres-ence of mutated CARD15 (P = 0.0147). Thus, this completely reconsti-tuted cell system supports the idea that PGN induces increased NF-κBactivation in the presence of mutated CARD15.

DISCUSSIONIt has been suggested that a deficient innate immune response due toa defective NF-κB pathway was responsible for the occurrence ofintestinal inflammation in patients with Crohn disease withCARD15 mutations1,9,17,18. However, this does not adequatelyexplain the overexpression of NF-κB and increased TH1 cytokines inthe inflamed tissues of patients with Crohn disease19–25. However,the presumption of an proinflammatory function of NOD2 camefrom experiments using NOD2-transfected HEK 293 cells that donot express TLR2 (refs. 32,33). Thus, those previous results wereunlikely to reflect the physiological function of NOD2. Here weshowed, using Card15–/– APCs, that NOD2 signaling inhibits apotentially inflammatory TH1 response mediated by TLR2. The keyfinding was that wild-type cells stimulated either by the TLR2 ago-nist PGN alone (which upon cellular uptake can lead to the genera-tion of the intracellular NOD2 agonist MDP) or by the syntheticTLR2 agonists Pam3Cys and Pam3CSK4 plus MDP had lower IL-12responses than did Card15–/– cells stimulated in the same way. Inaddition, MDP suppressed TLR2-induced IL-12 production in wild-type cells but not in Card15–/– cells in a dose-dependent way. Finally,this defect in Card15–/– cells was manifest in NOD2-deficient mice,which showed an abnormally high IL-12 response after systemicPGN injection. Thus, we propose that NOD2 affects Crohn diseaseby mediating an anti-inflammatory pathway through its ability todownmodulate the PGN-induced TLR2 signal.

Figure 8 CD11b+ cells from Card15–/– mice expressing transfected Card15 with a frameshift mutation show enhanced PGN-induced IL-12 production.Splenic CD11b+ cells obtained from Card15–/– mice were transfected with plasmid expressing wild-type Card15 or L980fs Card15 as a form of HVJenvelope and were cultured with LPS (1 µg/ml), MDP (10 µg/ml) or PGN (10 µg/ml). (a) Whole-cell extracts purified from Card15–/– CD11b+ cells wereanalyzed for expression of exogenous NOD2 protein 24 h after transfection. Cell lysates from HEK 293 T cells transfected with Card15 were used aspositive controls. (b,c) Culture supernatants collected at 48 h were analyzed by ELISA for secretion of IL-12p40 (b) and IL-12p70 (c). **, P < 0.01,compared with cytokine concentrations produced by wild-type CD11b+ cells transfected with empty vector (Empty). Results are presented as means ±s.d. Control concentrations of cytokines in cells without stimulation: IL-12p40, wild-type mice, 76 ± 23 pg/ml, and Card15–/– mice, 105 ± 16 pg/ml; IL-12p70, below the level of detection. (d) HT-29 cells were transfected with vectors expressing human CARD15 (10 ng) or L1007fs CARD15 (10 ng) withor without TLR2 (50 ng) in the presence of the luciferase reporter pNF-κB-Luc (50 ng) and pSV-β-galactosidase (10 ng). After transfection, cells werestimulated with MDP (10 µg/ml) or PGN (10 µg/ml). Results of NF-κB luciferase activity normalized to galactosidase activity are presented as means ±s.d. *, P < 0.05, between two groups.

a b c

d

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urei

mm

unol

ogy

Page 7: NOD2 is a negative regulator of Toll-like receptor 2–mediated T … · 2005. 5. 2. · NOD2 to act as an intracellular receptor for MDP suggests that NOD2 functions in activation

A RT I C L E S

806 VOLUME 5 NUMBER 8 AUGUST 2004 NATURE IMMUNOLOGY

Studies analyzing the mechanism of NOD2-mediated regulationof TLR2 responses showed that Card15–/– CD11b+ cells hadenhanced NF-κB activation, particularly with respect to c-Rel. Inaddition, inhibition of increased c-Rel expression using c-Rel siRNAin NOD2-deficient CD11b+ cells led to normalization of IL-12 pro-duction. This finding is consistent with reports that Rel–/– micedemonstrate resistance to autoimmune encephalomyelitis and colla-gen-induced arthritis because of the lack of IL-12 production29,34

and that upregulation of c-Rel expression is associated withincreased production of IL-12 by macrophages from an autoim-mune-prone mouse strain28. Moreover, transcription of mRNA forboth IL-12p40 (ref. 35) and IL-12p35 (ref. 36) in APCs is criticallydependent on c-Rel activity. Those previous findings relating to c-Rel, in conjunction with our own findings, suggest that one wayNOD2 signaling regulates TLR2-induced IL-12 production is byreducing c-Rel activation and translocation. In addition, theystrongly suggest that the relation of NOD2 to c-Rel expression andIL-12 responses is the molecular basis for the development of intesti-nal inflammation induced by NOD2 dysfunction.

It is not yet clear whether the mechanism of NOD2-mediated reg-ulation of c-Rel is dependent on or independent of the ability ofNOD2 to activate RICK (also known as CARDIAK), the CARD-containing serine-threonine kinase thought to link NOD2 to NF-κBactivation. Our inability to find evidence of a direct physical inter-action between TLR2 and NOD2 in immunoprecipitation studiesusing transfected HT-29 cells (data not shown) as well as previousstudies showing that RICK tranduces both NOD2 and TLR2 signalsto NF-κB11 suggest that RICK is indeed involved. However, if thelatter were true, RICK would have to transduce a MDP-inducedNOD2 signal that is positive for NF-κB activation and to induce aPGN-MDP–induced NOD2 signal that is negative for NF-κB activa-tion. One possibility to explain this paradox is that RICK differen-tially recruits an additional cofactor that affects NF-κB signalingdepending on whether it is activated by NOD2 or TLR2 and NOD2.

Card15–/– mice do not develop spontaneous intestinal inflamma-tion when reared in standard animal maintenance conditions.Nevertheless, there is good reason to believe that a defect in NOD2negative regulation is the main reason for the association ofCARD15 mutations with Crohn disease. First, IL-12 overproduc-tion and the subsequent effect of this on TH1 responses associatedwith NOD2 dysfunction is a chief immunological feature of Crohndisease–associated inflammation. Patients demonstrate increasedIL-12 secretion not only by lamina propria macrophages but alsoby lamina propria T cells, manifested by increased expression of thetranscription factor T-bet and IFN-γ secretion23. The idea that anincrease in the TH1 response is a basic cause of Crohn disease isnow supported by a study of patients treated with anti-IL-12 whoshowed amelioration of disease along with a reduction in IL-12 andIFN-γ secretion in the mucosa19. Second, studies of murine modelsof mucosal inflammation have provided abundant evidence thatantigens or other substances in the mucosal bacterial flora areindispensable in the induction and maintenance of mucosalinflammation20. Thus, it is logical to assume that components ofthe innate immune system, including TLR signaling, contribute tothe development of intestinal inflammation. Consistent with this,spontaneous enterocolitis associated with the overproduction ofIL-12 that develops in mice deficient in the transcriptional activa-tor STAT3 does not occur in the presence of TLR4 deficiency37,which suggests a requirement for TLR4 or another type of TLR-mediated signaling in the development of TH1 type colitis. In addi-tion, CpG administration leads to mucosal TLR9 signaling and

increased severity of dextran sodium sulfate–induced colitis38.Thus, unregulated TLR2 signaling occurring in response to theubiquitous PGNs expressed by mucosal microflora may be a dri-ving force in Crohn disease. Third, we have shown here that IL-12production by Card15–/– cells was normalized with a wild-typeCard15 construct but not with a Card15 construct with a mutationcausing a frameshift in the LRR.

We assume that intact NOD2 signaling prevents the developmentof Crohn disease by controlling PGN-mediated TH1 responses viaTLR2. However, support of this view will require direct studies of cellsfrom patients with Crohn disease to determine if inflammation isassociated with the putative abnormality in negative regulation ofPGN signaling by NOD2. In addition, further studies using Card15–/–

mice are required to determine the conditions necessary for theirexpression of disease. In a preliminary study of cells from two patientswith Crohn disease with a CARD15 mutation, we noted increased anddecreased IL-12 production by PGN- and MDP-stimulated mono-cytes, respectively, compared with those from healthy controls andpatients with Crohn disease with an intact CARD15 (T.W., A.K., G.Bouma and W.S., unpublished data). However, this result should beinterpreted cautiously, as some peripheral monocytes from patientsmay be preactivated because of the underlying inflammation andwould therefore be capable of producing increased amounts of TH1cytokines after stimulation.

As shown here, MDP by itself demonstrated only a weak abilityto induce the secretion of inflammatory cytokines compared withagonists that act directly on TLRs. However, MDP enhances IL-8(refs. 39,40) and TNF41 secretion by macrophages stimulated withPGN, lipoteichoic acid, LPS or CpG. In addition, TNF secretionwas similarly enhanced in wild-type splenocytes stimulated withPam3Cys or Pam3CSK4 in combination with MDP, whereas therewas no difference in Card15–/– cells (T.W., A.K. and W.S., unpub-lished data). This last finding suggests that the enhancing effect ofMDP is in fact acting via NOD2 and thus that NOD2 signaling canalso be positive for cytokine secretion in some conditions. In anycase, these data relate to the observation that Card15–/– mice do nothave an abnormality in TNF secretion, whereas patients withCrohn disease have high concentrations of TNF and are effectivelytreated by anti-TNF in many cases42. This inconsistency is all themore notable given that the abnormality in mouse NOD2-deficientcells is characterized by an increase in NF-κB activation, an intra-cellular event usually associated with increased secretion of TNF.Increased TNF secretion in patients with CARD15 mutations couldbe a secondary effect that does not ‘flow’ directly from the CARD15mutation. Instead, it may result from an innate and/or adaptiveimmune response to mucosal microflora components that occursafter inflammation is established. These responses do not necessar-ily involve PGNs and NOD2 signaling but could involve signalingthrough other TLRs. Finally, the lack of TNF overproduction inCard15–/– mice is probably not due to the abnormal NF-κB activa-tion in the absence of NOD2 function, because the qualitativechange in the NF-κB signal characteristic of NOD2 deficiency doesnot affect TNF production.

The function of NOD2 as a negative regulator of the TLR2-medi-ated cytokine response can also be applied to the treatment of patientswith Crohn disease with an intact CARD15. PGNs derived from mostGram-positive bacteria activate the NOD2 pathway only, whereasPGNs derived from Gram-negative bacteria stimulate both NOD1and NOD2 (ref. 43). Furthermore, excessive responses to intestinalbacterial flora have been proposed as one of the essential factorsresponsible for inflammatory bowel disease19,20. These findings,

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urei

mm

unol

ogy

Page 8: NOD2 is a negative regulator of Toll-like receptor 2–mediated T … · 2005. 5. 2. · NOD2 to act as an intracellular receptor for MDP suggests that NOD2 functions in activation

A RT I C L E S

NATURE IMMUNOLOGY VOLUME 5 NUMBER 8 AUGUST 2004 807

together with the results reported here, suggest that activation ofNOD2 signaling by Gram-positive but not Gram-negative bacteriamight decrease the inflammation of Crohn disease through downreg-ulation of the TH1 response. Thus, our study introduces the possibil-ity that patients with Crohn disease with an intact CARD15 can betreated by the introduction of Gram-positive bacteria into theirintestinal flora.

METHODSMice. Male wild-type and Card15–/– mice 6–10 weeks old26 were used. Animaluse adhered to National Institutes of Health Animal Care Guidelines.

Cell isolation and stimulation. Total splenocytes were isolated as described44.CD11b+ macrophages and CD4+ T cells were purified from total splenocytes bypositive selection with anti-mouse CD11b and anti-CD4 microbeads (MiltenyiBiotech). In cell depletion studies, samples were depleted of B cells, T cells ormacrophages by negative selection with anti-mouse B220, anti-CD4-CD8 andanti-CD11b microbeads, respectively. The purity of the cell subpopulation wasdetermined to be more than 90% by flow cytometry. Cells (1 × 106/ml)were stimulated for 48 h with LPS (1 µg/ml of ultrapure LPS lacking TLR2-stimulating activity; InvivoGen), PGN from Staphylococcus aureus(10 µg/ml; Fluka), Pam3Cys (500 ng/ml; InvivoGen), Pam3CSK4 (500 ng/ml;InvivoGen), dsRNA (25 µg/ml; InvivoGen), loxoribine (100 µM; InvivoGen),CpG (1 µM; InvivoGen) or MDP (10 µg/ml; Sigma) in RPMI 1640 mediumcontaining 10% FCS. Unless specified otherwise, the concentrations describedabove were used throughout the study. Culture supernatants were assayed for IFN-γ, IL-12p40, IL-18, IL-10 and TNF by enzyme-linked immunosorbent assay (ELISA) kits (Pharmingen) and for IL-12p70 with the Quantikinemouse IL-12p70 kit (R&D systems).

RT-PCR analysis of IL-12p40, p35 and IL-23p19 mRNA. Splenic CD11b+ cellswere stimulated with LPS or PGN for 5 h and then total RNA was isolated withthe RNeasy kit (Qiagen). A total of 100 ng of template RNA was used in the RT-PCR reaction with the OneStep RT-PCR kit (Qiagen). Primer sequences were asfollows: IL-12p40, 5′-CCTCAGAAGCTAACCATCTCCTG-3′ and 5′-ACATGTCACTGCCCGAGAGTCAG-3′; IL-12p35, 5′-CAATCACGCTACCTCCTCTTTTTG-3′ and 5′-CTCCCTCTTGTTGTGGAGAAGTC-3′; IL-23p19, 5′-ACCCACAAGGACTCAAGGACAAC-3′ and 5′-TGCCCTTCACGCAAAACAAAAC-3′;and glyceraldehyde phosphodehydrogenase (GAPDH), 5′-GGTGAAGGTCGGTGTGAACGGA-3′ and 5′-TGTTAGTGGGGTCTCGCTCCTG-3′. Annealingtemperature and cycle number were as follows: IL-12p40 and p35, 60 °C and 30 cycles; IL-23p19, 65 °C and 40 cycles; and GAPDH, 60 °C and 25 cycles.

Flow cytometry. Total splenocytes were stimulated for 24 h with LPS, PGNor dsRNA. Cell surface expression of IL-12Rβ1 and IL-12Rβ2 on CD4+

T cells was analyzed as described45. For staining of the IL-12Rβ2 chain, cellswere first treated with hamster monoclonal antibody (mAb) to IL-12Rβ2chain45. Then, cells were incubated with biotinylated anti-hamsterimmunoglobulin G (IgG; H+L; Jackson ImmunoResearch Laboratories)followed by phycoerythrin-conjugated streptavidin and fluorescein isothio-cyanate–conjugated mAb to anti-CD4 (Pharmingen). For staining of theIL-12Rβ1 chain, cells were incubated with biotinylated mAb to IL-12Rβ1(Pharmingen) followed by phycoerythrin-conjugated streptavidin and fluo-rescein isothiocyanate–conjugated mAb to CD4. For expression of cell sur-face TLR2, purified CD11b+ cells were stimulated with PGN (1 or 10 µg/ml),Pam3Cys (500 ng/ml) or Pam3CSK4 (500 ng/ml). Cells were incubatedwith biotinylated mAb to mouse TLR2 (eBioscience) followed by strepta-vidin-phycoerythrin. Nonspecific binding of antibodies was blocked by2.4G2 (Pharmingen), and dead cells were excluded by propidium iodidestaining. Analysis was done on a Becton Dickinson FACScan withCELLQuest II software.

Systemic challenge of wild-type and Card15–/– mice with PGN or LPS. Wild-type and Card15–/– mice were given 30 or 300 µg of LPS (InvivoGen) or PGN(Fluka) intravenously and then serum samples were collected 2, 4, and 6 hafter challenge time points. Serum concentrations of IL-12p40 and IL-12p70were determined by ELISA.

NF-κB activation assay. Splenic adherent cells were stimulated for 2 h withLPS, PGN or MDP. Preparation of nuclear extracts was done with theTransfector Extraction kit (Clontech) and binding activity of nuclear extractto NF-κB subunit, p50, p65 and c-Rel was measured with a MercuryTransfactor kit46 (Clontech). Nuclear extract (15 or 30 µg) was applied to eachwell coated with NF-κB consensus oligonucleotides and then wells were incu-bated with rabbit anti-p50, anti-p65 or anti-c-Rel followed by horseradishperoxidase–labeled anti-rabbit IgG. EMSAs were done as described47. Forbinding reactions, 5 µg nuclear extracts and 2 ng end-labeled NF-κB-bindingconsensus oligonucleotides (Santa Cruz Biotechnology) were incubated for 20 min at 25 °C in 15 µl binding buffer (20 mM HEPES, pH 7.9, 30 mM KCl, 4 mM MgCl2, 0.1 mM EDTA, 0.8 mM NaPi, 20% glycerol, 4 mM spermidineand 3 mg poly(dI:dC)). For supershift analyses, anti-p50, anti-p65 and anti-c-Rel (Santa Cruz Biotechnology) were used.

Assays with siRNA specific to c-Rel or p65. The siRNA assays specific to c-Relused the following target sequence of mouse c-Rel mRNA: 5′-AACAACCGGACAUACCCGUCU-3′ (ref. 28; Dharmacon). The siRNA targeting mousep65 mRNA was purchased from Santa Cruz Biotechnology. After 2 h of cul-ture, splenic CD11b+ cells were transfected with c-Rel siRNA with theTransIT-TKO transfection reagent28 (Mirus), followed by stimulation withLPS or PGN for 48 h. Whole-cell lysates and total RNA were prepared 6 h afterstimulation with PGN and were analyzed by immunoblot and RT-PCR.

Immunoblot analyses. Cells were lysed for 30 min on ice in lysis buffer (1%NP-40, 150 mM NaCl, 20 mM Tris, pH 7.5, and 2 mM EDTA) supplementedwith protease inhibitor ‘cocktails’ (Roche Molecular Biochemicals) and cen-trifuged at 12,000g for 30 min at 4 °C, and supernatants were collected. Thelysates were mixed with SDS sample buffer, incubated at 70 °C for 10 min,resolved by NUPAGE Novex Bis-Tris Gels (25 µg/well, Invitrogen) and trans-ferred to nitrocellulose membranes (Amersham Pharmacia Biotech). Theblotted membranes were blocked with 5% skim milk and 0.1% Tween 20 inTris-buffered saline, washed and then incubated with anti-c-Rel, anti-p65,anti-actin (Santa Cruz Biotechnology), or NOD2 polyclonal rabbit antiserum(Cayman) followed by incubation with horseradish peroxidase–conjugatedanti-goat IgG or anti-rabbit IgG (Zymed). Then, the membranes were devel-oped with the SuperSignal West Pico Chemiluminescent Substrate (PierceChemical) and exposed to X-ray film.

Preparation of HVJ envelope vector. HVJ envelope vector was prepared asdescribed48. The virus suspension (25,600 hemagglutinating units; AnGesMG) was inactivated by ultraviolet irradiation and was centrifuged at 10,000gfor 15 min at 4 °C. Pellets of HVJ envelope vector were mixed with 37.5 µl ofprotamine sulfate (1 mg/ml) followed by incubation for 10 min on ice. Then,plasmid DNA (100 µg/100 µl) and 13.8 µl of 0.3% Triton-X were added, fol-lowed by incubation for 15 min on ice. After centrifugation at 15,000g for 15 min, HVJ envelope–plasmid DNA was suspended in 500 µl of PBS contain-ing 72 µg of protamine sulfate.

Transfection of plasmids expressing mouse Card15. The pcDNA4HisMaxplasmid vector expressing mouse NOD2 cDNA was provided by J.T. Rosenbaum(Casey Eye Institute, Oregon Health and Science University, Portland,Oregon)49. The mutant plasmid for L980fs was generated with theQuickChange XL Site-Directed Mutagenesis kit (Stratagene). The authenticityof the construct was confirmed by DNA sequencing. Splenic CD11b+ cells fromCard15–/– mice were transfected with pcDNA4HisMax plasmid (empty) or plas-mid expressing wild-type or L980fs Card15 as a form of HVJ envelope (6 µgDNA per six-well plate; 2 µg per 24-well plate) followed by stimulation withPGN or MDP for 48 h. CD11b+ cells from wild-type mice were transfected withempty plasmid–HVJ envelope. At 24 h after transfection, cell lysates were pre-pared and analyzed for expression of exogenous NOD2 protein by immunoblotwith NOD2 antiserum. HEK 293T cells (ATCC) were transfected with empty,wild-type or L980fs Card15 vector using the Trans-ITLT1 reagent (Mirus), andlysates from these cells were used as controls for exogenous expression of NOD2.

Luciferase assay. The pUNO plasmid expressing human CARD15 or TLR2cDNA was obtained from InvivoGen. The mutant plasmid for L1007fsinsCwas generated with the QuickChange XL Site-Directed Mutagenesis kit, and

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urei

mm

unol

ogy

Page 9: NOD2 is a negative regulator of Toll-like receptor 2–mediated T … · 2005. 5. 2. · NOD2 to act as an intracellular receptor for MDP suggests that NOD2 functions in activation

A RT I C L E S

808 VOLUME 5 NUMBER 8 AUGUST 2004 NATURE IMMUNOLOGY

the authenticity of the construct was confirmed by DNA sequencing. HT-29cells (1.5 × 105 cells/ml; ATCC) were transiently transfected with the luciferasereporter plasmid pNF-κB-Luc containing four κB-binding sites (Clontech)and the pSV-β-galactosidase vector (Promega), together with a plasmidexpressing CARD15, L1007fs CARD15 and/or TLR2, using the Trans-ITLT1reagent. After overnight incubation in serum-free medium, cells were stimu-lated with MDP or PGN for 5 h and then cell lysates were analyzed forluciferase activity (Promega) and galactosidase activity (Applied Biosystems)for normalization.

Statistical analyses. Student’s t-test was used to evaluate the significance of thedifferences. Statistical analysis was done with the StatView v.4.5 program(Abacus Concepts). A value of P < 0.05 was considered statistically significant.

ACKNOWLEDGMENTSWe thank J.T. Rosenbaum and M.P. Davey (Casey Eye Institute, Oregon Health andScience University) for providing us with the plasmid expressing mouse Card15;and C. Ma and S. Fichtner (Laboratory of Host Defenses, National Institute ofAllergy and Infectious Diseases) for comments and technical assistance. Supportedin part by the Cancer Center CORE (P30 CA21765) and the American LebaneseAssociated Charities.

COMPETING INTERESTS STATEMENTThe authors declare that they have no competing financial interests.

Received 14 April; accepted 25 May 2004Published online at http://www.nature.com/natureimmunology/

1. Ogura, Y. et al. A frameshift mutation in NOD2 associated with susceptibility toCrohn’s disease. Nature 411, 603–606 (2001).

2. Hugot, J.P. et al. Association of NOD2 leucine-rich repeat variants with susceptibil-ity to Crohn’s disease. Nature 411, 599–603 (2001).

3. Cuthbert, A.P. et al. The contribution of NOD2 gene mutations to the risk and site ofdisease in inflammatory bowel disease. Gastroenterology 122, 867–874 (2002).

4. Hampe, J. et al. Association between insertion mutation in NOD2 gene and Crohn’sdisease in German and British populations. Lancet 357, 1925–1928 (2001).

5. Inohara, N. & Nunez, G. NODs: intracellular proteins involved in inflammation andapoptosis. Nat. Rev. Immunol. 3, 371–382 (2003).

6. Chamaillard, M., Girardin, S.E., Viala, J. & Philpott, D.J. Nods, Nalps and Naip:intracellular regulators of bacterial-induced inflammation. Cell. Microbiol. 5,581–592 (2003).

7. Harton, J.A., Linhoff, M.W., Zhang, J. & Ting, J.P. Cutting edge: CATERPILLER: alarge family of mammalian genes containing CARD, pyrin, nucleotide-binding, andleucine-rich repeat domains. J. Immunol. 169, 4088–4093 (2002).

8. Gutierrez, O. et al. Induction of Nod2 in myelomonocytic and intestinal epithelialcells via nuclear factor-κB activation. J. Biol. Chem. 277, 41701–41705 (2002).

9. Inohara, N. et al. Host recognition of bacterial muramyl dipeptide mediated throughNOD2. Implications for Crohn’s disease. J. Biol. Chem. 278, 5509–5512 (2003).

10. Girardin, S.E. et al. Nod2 is a general sensor of peptidoglycan through muramyldipeptide (MDP) detection. J. Biol. Chem. 278, 8869–8872 (2003).

11. Kobayashi, K. et al. RICK/Rip2/CARDIAK mediates signalling for receptors of theinnate and adaptive immune systems. Nature 416, 194–199 (2002).

12. Ogura, Y. et al. Nod2, a Nod1/Apaf-1 family member that is restricted to monocytesand activates NF-κB. J. Biol. Chem. 276, 4812–4818 (2001).

13. Girardin, S.E. et al. Nod1 detects a unique muropeptide from gram-negative bacter-ial peptidoglycan. Science 300, 1584–1587 (2003).

14. Chamaillard, M. et al. An essential role for NOD1 in host recognition of bacterialpeptidoglycan containing diaminopimelic acid. Nat. Immunol. 4, 702–707 (2003).

15. Akira, S., Takeda, K. & Kaisho, T. Toll-like receptors: critical proteins linking innateand acquired immunity. Nat. Immunol. 2, 675–680 (2001).

16. Akira, S. Toll-like receptor signaling. J. Biol. Chem. 278, 38105–38108 (2003).17. Chamaillard, M. et al. Gene-environment interaction modulated by allelic hetero-

geneity in inflammatory diseases. Proc. Natl. Acad. Sci. USA 100, 3455–3460(2003).

18. Bonen, D.K. et al. Crohn’s disease-associated NOD2 variants share a signalingdefect in response to lipopolysaccharide and peptidoglycan. Gastroenterology 124,140–146 (2003).

19. Bouma, G. & Strober, W. The immunological and genetic basis of inflammatorybowel disease. Nat. Rev. Immunol. 3, 521–533 (2003).

20. Strober, W., Fuss, I.J. & Blumberg, R.S. The immunology of mucosal models ofinflammation. Annu. Rev. Immunol. 20, 495–549 (2002).

21. Monteleone, G. et al. Interleukin 12 is expressed and actively released by Crohn’sdisease intestinal lamina propria mononuclear cells. Gastroenterology 112,1169–1178 (1997).

22. Kanai, T. et al. Interleukin 18 is a potent proliferative factor for intestinal mucosallymphocytes in Crohn’s disease. Gastroenterology 119, 1514–1523 (2000).

23. Neurath, M.F. et al. The transcription factor T-bet regulates mucosal T cell activationin experimental colitis and Crohn’s disease. J. Exp. Med. 195, 1129–1143 (2002).

24. Fuss, I.J. et al. Disparate CD4+ lamina propria (LP) lymphokine secretion profiles ininflammatory bowel disease. Crohn’s disease LP cells manifest increased secretionof IFN-γ, whereas ulcerative colitis LP cells manifest increased secretion of IL-5. J. Immunol. 157, 1261–1270 (1996).

25. Parronchi, P. et al. Type 1 T-helper cell predominance and interleukin-12 expressionin the gut of patients with Crohn’s disease. Am. J. Pathol. 150, 823–832 (1997).

26. Pauleau, A.L. & Murray, P.J. Role of nod2 in the response of macrophages to toll-likereceptor agonists. Mol. Cell. Biol. 23, 7531–7539 (2003).

27. Nakayama, K. et al. Involvement of IRAK-M in peptidoglycan-induced tolerance inmacrophages. J.Biol. Chem. 279, 6629–6634 (2004).

28. Liu, J. & Beller, D.I. Distinct pathways for NF-κB regulation are associated withaberrant macrophage IL-12 production in lupus- and diabetes-prone mouse strains. J. Immunol. 170, 4489–4496 (2003).

29. Hilliard, B.A. et al. Critical roles of c-Rel in autoimmune inflammation and helper T cell differentiation. J. Clin. Invest. 110, 843–850 (2002).

30. Hisamatsu, T. et al. CARD15/NOD2 functions as an antibacterial factor in humanintestinal epithelial cells. Gastroenterology 124, 993–1000 (2003).

31. Cario, E. et al. Lipopolysaccharide activates distinct signaling pathways in intestinalepithelial cell lines expressing Toll-like receptors. J. Immunol. 164, 966–972 (2000).

32. Smith, M.F., Jr. et al. Toll-like receptor (TLR) 2 and TLR5, but not TLR4, arerequired for Helicobacter pylori-induced NF-κB activation and chemokine expres-sion by epithelial cells. J. Biol. Chem. 278, 32552–32560 (2003).

33. Fisette, P.L., Ram, S., Andersen, J.M., Guo, W. & Ingalls, R.R. The Lip lipoproteinfrom Neisseria gonorrhoeae stimulates cytokine release and NF-κB activation inepithelial cells in a Toll-like receptor 2-dependent manner. J. Biol. Chem. 278,46252–46260 (2003).

34. Campbell, I.K., Gerondakis, S., O’Donnell, K. & Wicks, I.P. Distinct roles for the NF-κB1 (p50) and c-Rel transcription factors in inflammatory arthritis. J. Clin. Invest.105, 1799–1806 (2000).

35. Sanjabi, S., Hoffmann, A., Liou, H.C., Baltimore, D. & Smale, S.T. Selective require-ment for c-Rel during IL-12 P40 gene induction in macrophages. Proc. Natl. Acad.Sci. USA 97, 12705–12710 (2000).

36. Grumont, R. et al. c-Rel regulates interleukin 12 p70 expression in CD8+ dendritic cellsby specifically inducing p35 gene transcription. J. Exp. Med. 194, 1021–1032 (2001).

37. Kobayashi, M. et al. Toll-like receptor-dependent production of IL-12p40 causeschronic enterocolitis in myeloid cell-specific Stat3-deficient mice. J. Clin. Invest.111, 1297–1308 (2003).

38. Obermeier, F. et al. CpG motifs of bacterial DNA exacerbate colitis of dextran sulfatesodium-treated mice. Eur. J. Immunol. 32, 2084–2092 (2002).

39. Yang, S. et al. Synergistic effect of muramyldipeptide with lipopolysaccharide orlipoteichoic acid to induce inflammatory cytokines in human monocytic cells in cul-ture. Infect. Immun. 69, 2045–2053 (2001).

40. Takada, H., Yokoyama, S. & Yang, S. Enhancement of endotoxin activity bymuramyldipeptide. J. Endotoxin. Res. 8, 337–342 (2002).

41. Wolfert, M.A., Murray, T.F., Boons, G.J. & Moore, J.N. The origin of the synergisticeffect of muramyl dipeptide with endotoxin and peptidoglycan. J. Biol. Chem. 277,39179–39186 (2002).

42. Targan, S.R. et al. A short-term study of chimeric monoclonal antibody cA2 to tumornecrosis factor α for Crohn’s disease. Crohn’s Disease cA2 Study Group. N. Engl. J.Med. 337, 1029–1035 (1997).

43. Girardin, S.E., Hugot, J.P. & Sansonetti, P.J. Lessons from Nod2 studies: towards a linkbetween Crohn’s disease and bacterial sensing. Trends. Immunol. 24, 652–658 (2003).

44. Watanabe, T. et al. Administration of an antigen at a high dose generates regulatoryCD4+ T cells expressing CD95 ligand and secreting IL-4 in the liver. J. Immunol.168, 2188–2199 (2002).

45. Nishikomori, R., Ehrhardt, R.O. & Strober, W. T helper type 2 cell differentiationoccurs in the presence of interleukin 12 receptor β2 chain expression and signaling.J. Exp. Med. 191, 847–858 (2000).

46. O’Sullivan, B.J. & Thomas, R. CD40 ligation conditions dendritic cell antigen-pre-senting function through sustained activation of NF-κB. J. Immunol. 168,5491–5498 (2002).

47. Kitani, A. et al. Transforming growth factor (TGF)-β1-producing regulatory T cellsinduce Smad-mediated interleukin 10 secretion that facilitates coordinatedimmunoregulatory activity and amelioration of TGF-β1-mediated fibrosis. J. Exp.Med. 198, 1179–1188 (2003).

48. Shimamura, M. et al. HVJ-envelope vector for gene transfer into central nervous sys-tem. Biochem. Biophys. Res. Commun. 300, 464–471 (2003).

49. Iwanaga, Y. et al. Cloning, sequencing and expression analysis of the mouseNOD2/CARD15 gene. Inflamm. Res. 52, 272–276 (2003).

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urei

mm

unol

ogy


Recommended