neddylation pathway regulates t-cell function by targeting an … · neddylation pathway regulates...

6
Neddylation pathway regulates T-cell function by targeting an adaptor protein Shc and a protein kinase Erk signaling Hyung-seung Jin a , Lujian Liao b , Yoon Park a , and Yun-Cai Liu a,1 a Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037 and b Shanghai Institute of Biochemistry and Cell Biology, Shanghai 200031, China Edited by Michael Karin, University of California at San Diego School of Medicine, La Jolla, CA, and approved November 28, 2012 (received for review August 9, 2012) NEDD8 (neural precursor cell expressed, developmentally down- regulated 8) is a ubiquitin-like molecule whose action on modify- ing protein substrates is critical in various cellular functions but whose importance in the immune system is not well understood. Here we investigated the role of protein neddylation in regulating T-cell function using an in vivo knockdown technique. We found that reduced expression of Ubc12 in CD4 + T cells led to impaired T-cell receptor/CD28-induced proliferation and cytokine production both in vitro and in vivo, accompanied by reduced Erk activation. These ndings were recapitulated by treatment with MLN4924, an inhibitor of NEDD8-activating enzyme. Furthermore, Shc, an adap- tor molecule between antigen receptors and the Ras/Erk pathway, was identied as a target for neddylation. Importantly, mice adop- tively transferred with Ubc12 knockdown CD4 + T cells showed markedly ameliorated allergic responses. This study thus identies an important role for protein neddylation in T-cell function, which may serve as a therapeutic target for inammatory diseases. allergy | signal transduction | posttranslational modication E ngagement of T-cell antigen receptor (TCR) and cos- timulatory molecules leads to the activation of CD4 + T cells through several signaling pathways, which ultimately induces proliferation, cytokine production, and differentiation into dif- ferent subsets of T helper type (Th) cells and memory cells (1). Notably, posttranslational modication by ubiquitin and ubiq- uitin-like proteins (UBLs) has emerged as a critical mechanism regulating T-cell function (2, 3). UBLs compose a diverse group of evolutionarily conserved small proteins, and attachment of different UBLs to a target has different biological consequences (4). Among the UBLs, NEDD8 (neural precursor cell expressed, developmentally down-regulated 8) is covalently attached to lysines in substrate proteins by a series of enzymatic reactions similar to those involved in ubiquitination, including the E1 (Nae1/Uba3) and E2 (Ubc12) reactions (5). The well-charac- terized substrates of NEDD8 modication are the cullin subunits of cullin-RING ubiquitin E3 ligases, and this modication of cullins is critical for the transfer of ubiquitin from recruited E2- ubiquitin to the substrate (6, 7). The importance of this pathway was further underscored by the recent development of a phar- macologic inhibitor of Nae1, MLN4924, which inhibits cullin neddylation, resulting in substrate accumulation and cancer cell apoptosis by the deregulation of S-phase DNA synthesis, and offers great promise for the treatment of cancer (810). In the present study, we investigated whether the NEDD8 pathway is involved in CD4 + T-cell function. Using siRNA- mediated depletion of the NEDD8 system in vivo and the NEDD8-activating enzyme inhibitor MLN4924, we found that the NEDD8 pathway is required for TCR-induced proliferation and activation. Interestingly, our results demonstrate that Erk activation by TCR stimulation requires an intact neddylation system. Differentiation of CD4 + T cells from Ubc12 knockdown bone marrow chimeric mice into Th1 and Th2 subsets was markedly decreased. Mice transferred with Ubc12 knockdown CD4 + T cells exhibited a deciency in the ability to develop T-celldriven airway inammation. Thus, the NEDD8 pathway appears to play an important role in CD4 + T-cellmediated inammatory responses. Results and Discussion NEDD8 Pathway Is Required for CD4 + T-Cell Function. To explore the function of the NEDD8 pathway in the immune system, partic- ularly in CD4 + T cells, we generated retroviral vectors expressing both GFP and an shRNA targeting Ubc12. Bone marrow cells isolated from SJL B6 mice (CD45.1 + ) were transduced with the retrovirus bearing Ubc12 shRNA or control shRNA, then transplanted into lethally irradiated C57BL/6 mice (CD45.2 + ). After 8 wk, newly reconstituted GFP + CD4 + T cells were sorted from the spleen and lymph nodes of bone marrow chimeric mice. Western blot analysis showed a signicant decrease in Ubc12 protein expression in the sorted GFP + T-cell population (Fig. 1A). We next analyzed T-cell development in the thymuses of Ubc12 knockdown chimeric mice. Decreased expression of Ubc12 had no obvious effect on the development of CD4 + , CD8 + , or CD4 + CD8 + thymocytes, and similarly, the proportion of CD4 + and CD8 + T cells in the spleens of these mice was comparable to that in control mice (Fig. S1 A and B). We quantied memory and naive T cells on the basis of surface ex- pression of the activation markers CD44 and CD62L. Compared with control mice, the Ubc12 knockdown chimeric mice had a signicantly lower frequency of memory CD4 + T cells with a concomitantly higher frequency of naive CD4 + T cells (Fig. S1C). To assess the function of Ubc12 in T-cell proliferation, we stimulated control and Ubc12 knockdown naïve CD4 + T cells in vitro with anti-CD3 and anti-CD28. Proliferation of the stimu- lated knockdown T cells was substantially reduced as measured by 3 H-thymidine incorporation (Fig. 1A). Staining with 5-ethy- nyl-2-deoxyuridine (EdU) and 7-amino-actinomycin D (7-ADD) revealed that Ubc12 knockdown T cells were blocked in the G0/G1 phase of the cell cycle and did not progress efciently through the S phase (Fig. 1B). Analysis of individual cell division of violet dye-labeled CD4 + T cells by ow cytometry showed that Ubc12 knockdown T cells divided much more slowly than control T cells (Fig. 1C) and also failed to up-regulate the activation markers CD69 and CD25 to the same extent as control cells (Fig. 1D). Evaluation of cell death by a 7-AAD exclusion assay Author contributions: H.-s.J., Y.P., and Y.-C.L. designed research; H.-s.J., L.L., and Y.P. performed research; L.L. contributed new reagents/analytic tools; H.-s.J., L.L., Y.P., and Y.-C.L. analyzed data; and H.-s.J. and Y.-C.L. wrote the paper. The authors declare no conict of interest. This article is a PNAS Direct Submission. 1 To whom correspondence should be addressed. E-mail: [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1213819110/-/DCSupplemental. 624629 | PNAS | January 8, 2013 | vol. 110 | no. 2 www.pnas.org/cgi/doi/10.1073/pnas.1213819110 Downloaded by guest on May 30, 2021

Upload: others

Post on 25-Jan-2021

8 views

Category:

Documents


0 download

TRANSCRIPT

  • Neddylation pathway regulates T-cell functionby targeting an adaptor protein Shc and a proteinkinase Erk signalingHyung-seung Jina, Lujian Liaob, Yoon Parka, and Yun-Cai Liua,1

    aDivision of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037 and bShanghai Institute of Biochemistry and Cell Biology,Shanghai 200031, China

    Edited by Michael Karin, University of California at San Diego School of Medicine, La Jolla, CA, and approved November 28, 2012 (received for reviewAugust 9, 2012)

    NEDD8 (neural precursor cell expressed, developmentally down-regulated 8) is a ubiquitin-like molecule whose action on modify-ing protein substrates is critical in various cellular functions butwhose importance in the immune system is not well understood.Here we investigated the role of protein neddylation in regulatingT-cell function using an in vivo knockdown technique. We foundthat reduced expression of Ubc12 in CD4+ T cells led to impairedT-cell receptor/CD28-induced proliferation and cytokine productionboth in vitro and in vivo, accompanied by reduced Erk activation.These findings were recapitulated by treatment with MLN4924, aninhibitor of NEDD8-activating enzyme. Furthermore, Shc, an adap-tor molecule between antigen receptors and the Ras/Erk pathway,was identified as a target for neddylation. Importantly, mice adop-tively transferred with Ubc12 knockdown CD4+ T cells showedmarkedly ameliorated allergic responses. This study thus identifiesan important role for protein neddylation in T-cell function, whichmay serve as a therapeutic target for inflammatory diseases.

    allergy | signal transduction | posttranslational modification

    Engagement of T-cell antigen receptor (TCR) and cos-timulatory molecules leads to the activation of CD4+ T cellsthrough several signaling pathways, which ultimately inducesproliferation, cytokine production, and differentiation into dif-ferent subsets of T helper type (Th) cells and memory cells (1).Notably, posttranslational modification by ubiquitin and ubiq-uitin-like proteins (UBLs) has emerged as a critical mechanismregulating T-cell function (2, 3). UBLs compose a diverse groupof evolutionarily conserved small proteins, and attachment ofdifferent UBLs to a target has different biological consequences(4). Among the UBLs, NEDD8 (neural precursor cell expressed,developmentally down-regulated 8) is covalently attached tolysines in substrate proteins by a series of enzymatic reactionssimilar to those involved in ubiquitination, including the E1(Nae1/Uba3) and E2 (Ubc12) reactions (5). The well-charac-terized substrates of NEDD8 modification are the cullin subunitsof cullin-RING ubiquitin E3 ligases, and this modification ofcullins is critical for the transfer of ubiquitin from recruited E2-ubiquitin to the substrate (6, 7). The importance of this pathwaywas further underscored by the recent development of a phar-macologic inhibitor of Nae1, MLN4924, which inhibits cullinneddylation, resulting in substrate accumulation and cancer cellapoptosis by the deregulation of S-phase DNA synthesis, andoffers great promise for the treatment of cancer (8–10).In the present study, we investigated whether the NEDD8

    pathway is involved in CD4+ T-cell function. Using siRNA-mediated depletion of the NEDD8 system in vivo and theNEDD8-activating enzyme inhibitor MLN4924, we found thatthe NEDD8 pathway is required for TCR-induced proliferationand activation. Interestingly, our results demonstrate that Erkactivation by TCR stimulation requires an intact neddylationsystem. Differentiation of CD4+ T cells from Ubc12 knockdownbone marrow chimeric mice into Th1 and Th2 subsets was

    markedly decreased. Mice transferred with Ubc12 knockdownCD4+ T cells exhibited a deficiency in the ability to developT-cell–driven airway inflammation. Thus, the NEDD8 pathwayappears to play an important role in CD4+ T-cell–mediatedinflammatory responses.

    Results and DiscussionNEDD8 Pathway Is Required for CD4+ T-Cell Function. To explore thefunction of the NEDD8 pathway in the immune system, partic-ularly in CD4+ T cells, we generated retroviral vectors expressingboth GFP and an shRNA targeting Ubc12. Bone marrow cellsisolated from SJL B6 mice (CD45.1+) were transduced with theretrovirus bearing Ubc12 shRNA or control shRNA, thentransplanted into lethally irradiated C57BL/6 mice (CD45.2+).After 8 wk, newly reconstituted GFP+CD4+ T cells were sortedfrom the spleen and lymph nodes of bone marrow chimeric mice.Western blot analysis showed a significant decrease in Ubc12protein expression in the sortedGFP+T-cell population (Fig. 1A).We next analyzed T-cell development in the thymuses of

    Ubc12 knockdown chimeric mice. Decreased expression ofUbc12 had no obvious effect on the development of CD4+,CD8+, or CD4+CD8+ thymocytes, and similarly, the proportionof CD4+ and CD8+ T cells in the spleens of these mice wascomparable to that in control mice (Fig. S1 A and B). Wequantified memory and naive T cells on the basis of surface ex-pression of the activation markers CD44 and CD62L. Comparedwith control mice, the Ubc12 knockdown chimeric mice hada significantly lower frequency of memory CD4+ T cells with aconcomitantly higher frequency of naive CD4+ T cells (Fig. S1C).To assess the function of Ubc12 in T-cell proliferation, we

    stimulated control and Ubc12 knockdown naïve CD4+ T cells invitro with anti-CD3 and anti-CD28. Proliferation of the stimu-lated knockdown T cells was substantially reduced as measuredby 3H-thymidine incorporation (Fig. 1A). Staining with 5-ethy-nyl-2′-deoxyuridine (EdU) and 7-amino-actinomycin D (7-ADD)revealed that Ubc12 knockdown T cells were blocked in theG0/G1 phase of the cell cycle and did not progress efficientlythrough the S phase (Fig. 1B). Analysis of individual cell divisionof violet dye-labeled CD4+ T cells by flow cytometry showed thatUbc12 knockdown T cells divided much more slowly than controlT cells (Fig. 1C) and also failed to up-regulate the activationmarkers CD69 and CD25 to the same extent as control cells(Fig. 1D). Evaluation of cell death by a 7-AAD exclusion assay

    Author contributions: H.-s.J., Y.P., and Y.-C.L. designed research; H.-s.J., L.L., and Y.P.performed research; L.L. contributed new reagents/analytic tools; H.-s.J., L.L., Y.P., andY.-C.L. analyzed data; and H.-s.J. and Y.-C.L. wrote the paper.

    The authors declare no conflict of interest.

    This article is a PNAS Direct Submission.1To whom correspondence should be addressed. E-mail: [email protected].

    This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1213819110/-/DCSupplemental.

    624–629 | PNAS | January 8, 2013 | vol. 110 | no. 2 www.pnas.org/cgi/doi/10.1073/pnas.1213819110

    Dow

    nloa

    ded

    by g

    uest

    on

    May

    30,

    202

    1

    http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1213819110/-/DCSupplemental/pnas.201213819SI.pdf?targetid=nameddest=SF1http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1213819110/-/DCSupplemental/pnas.201213819SI.pdf?targetid=nameddest=SF1mailto:[email protected]://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1213819110/-/DCSupplementalhttp://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1213819110/-/DCSupplementalwww.pnas.org/cgi/doi/10.1073/pnas.1213819110

  • showed similar 7-AAD staining in control and Ubc12 knockdownT cells after 24 h of TCR stimulation (Fig. S2), suggesting thatUbc12 knockdown does not affect the survival of CD4+ T cellsin response to TCR activation. Taken together, these resultsdemonstrate a critical role for the NEDD8 pathway in the pos-itive regulation of CD4+ T-cell activation.To further test whether other components of the NEDD8

    pathway also affect T-cell function, we designed a single retro-viral vector bearing three shRNA cassettes targeting the ned-dylation components of Nae1, Ubc12, and NEDD8 (shTriple) orindividually (Fig. S3A). The knockdown efficiencies of various

    hairpin constructs were measured by Western blot analysis (Fig.S3B). Inhibition of the NEDD8 pathway by the shRNA-medi-ated simultaneous knockdown of the neddylation componentsalso led to impaired proliferation and division of CD4+ T cells(Fig. S3 C and D). In addition, IL-2 production was substantiallyreduced in the triple-knockdown T cells (Fig. S3E). Further-more, Nae1-only knockdown CD4+ T cells also displayed a de-crease in proliferation and IL-2 production in response to TCRactivation (Fig. S3 F and G).

    Defective T-Cell Differentiation by Ubc12 Knockdown. We next in-vestigated the function of the NEDD8 pathway in TCR-induced

    D

    CD69 CD25

    shUbc12ctl

    IL-2

    (pg

    /ml)

    0

    100

    200

    300

    400

    500

    CD3 4µg +CD28

    CD3 1µg +CD28

    E

    0

    50

    100

    150

    200

    250

    IFN

    - (

    pg/m

    l)

    CD3 4µg +CD28

    CD3 1µg +CD28

    shUbc12ctl

    B ctl shUbc12

    DNA content (7-AAD)

    7.4

    84.5 96.3

    0.67

    EdU

    C ctl shUbc12

    Cell Trace

    F

    IFN

    -IL

    -2

    GFP

    ctl shUbc12

    A

    0

    20

    40

    60

    80

    shUbc12ctl

    [H3 ]

    Thy

    mid

    ine

    ctl shUbc1

    2

    CD3 (µg/ml) 2 5 10 2 5 10 0 CD28 (µg/ml) 0 0 0 2 2 2 2

    (x102)

    ctl shUbc12

    IFN

    -

    TH1

    TH2

    IL-4

    G

    Fig. 1. Ubc12 is required for TCR-induced proliferation and activation. (A) Immunoblot analysis of Ubc12 was performed in sorted GFP+ control (ctl) or GFP+

    shUbc12 CD4+ T cells (Inset). Peripheral naïve CD4+ T cells sorted from shRNA expressing chimeric mice were stimulated with various concentration of anti-CD3and anti-CD28 mAb for 3 d and then pulsed for an additional 8 h with 3H-thymidine. Cell proliferation was measured by 3H-thymidine uptake. (B) Cell cycleanalysis of control (Left) and shUbc12 CD4+ (Right) T cells. The cells were activated for 23 h with anti-CD3/CD28 mAbs, then pulsed for 1 h with EdU andanalyzed by flow cytometry after staining for EdU and 7-AAD. (C) Control (Left) and shUbc12 CD4+ (Right) violet-labeled CD4+ T cells were stimulated withanti-CD3/CD28 mAbs for 2 d, after which cell division was analyzed by flow cytometry. (D) Flow cytometry for cell surface expression of CD69 (Left) and CD25(Right) in sorted control and shUbc12 CD4+ T cells stimulated for 16 h with anti-CD3/CD28 mAbs. The results are representative of three repeated experiments.(E) IL-2 (Left) and IFN-γ (Right) production in control and shUbc12 CD4+ T cells stimulated for 48 h with anti-CD3 and anti-CD28, measured by ELISA. (F) Control(Left) and Ubc12 shRNA-expressing (Right) OT-II TCR transgenic mice were generated by reconstitution of retrovirally transduced bone marrow cells from OT-IImice into lethally irradiated recipient mice (n = 3). After 8 w, isolated CD4+ T cells from OT-II control or OT-II Ubc12 knockdown chimeric mice were adoptivelytransferred into C57BL/6J recipient mice, followed by immunization with OVA plus CFA. At 5 d after immunization, splenocytes and lymph node cells werestimulated with OVA323–339 peptide for 24 h and analyzed by flow cytometry using anti–IL-2 and anti–IFN-γ antibodies. (G) Sorted naïve CD4+ T cells werestimulated with anti-CD3/CD28 mAbs and differentiated in vitro under Th1-inducing (Upper) or Th2-inducing (Lower) conditions. Results are from threerepeated experiments.

    Jin et al. PNAS | January 8, 2013 | vol. 110 | no. 2 | 625

    IMMUNOLO

    GY

    Dow

    nloa

    ded

    by g

    uest

    on

    May

    30,

    202

    1

    http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1213819110/-/DCSupplemental/pnas.201213819SI.pdf?targetid=nameddest=SF2http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1213819110/-/DCSupplemental/pnas.201213819SI.pdf?targetid=nameddest=SF3http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1213819110/-/DCSupplemental/pnas.201213819SI.pdf?targetid=nameddest=SF3http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1213819110/-/DCSupplemental/pnas.201213819SI.pdf?targetid=nameddest=SF3http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1213819110/-/DCSupplemental/pnas.201213819SI.pdf?targetid=nameddest=SF3http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1213819110/-/DCSupplemental/pnas.201213819SI.pdf?targetid=nameddest=SF3http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1213819110/-/DCSupplemental/pnas.201213819SI.pdf?targetid=nameddest=SF3

  • cytokine production. We found that the secretion of IL-2 andIFN-γ was markedly impaired in both shUbc12- and shTriple-expressing cells compared with control cells (Fig. 1E and Fig. S3E).We further examined the in vivo function of the neddylationpathway in CD4+ T-cell–mediated immune responses using theOT-II transgenic TCR adoptive transfer system. For this, puri-fied CD4+ T cells from OT-II control or OT-II Ubc12 knock-down chimeric mice were adoptively transferred into C57BL/6Jrecipient mice. These mice were then immunized with ovalbumin(OVA) emulsified in complete Freund’s adjuvant (CFA) at 1 dafter adoptive transfer. Splenocytes obtained from OVA-immu-nized mice were cultured in the presence of OVA, and IFN-γand IL-2 production was analyzed by intracellular cytokinestaining. Splenic CD4+ T cells from the mice transferred withUbc12 knockdown OT-II CD4+ T cells exhibited impaired abilityto produce IFN-γ and IL-2 in response to a secondary antigenchallenge in vitro (Fig. 1F). These data suggest that the neddy-lation pathway is involved in antigen-driven cytokine productionby CD4+ T cells.We then examined whether Ubc12 knockdown affects the

    differentiation of CD4+ T cells into effector Th cell subsets. Westimulated naïve CD4+ T cells from control and Ubc12 knock-down chimeric mice with anti-CD3 and anti-CD28 mAbs, cul-tured them under either Th1- or Th2-polarizing conditions, andthen analyzed IFN-γ and IL-4 production. Ubc12 knockdownCD4+ T cells produced less IFN-γ and IL-4 compared with

    control cells under Th1 and Th2 culture conditions, respectively(Fig. 1G), indicating that the NEDD8 pathway is required forefficient Th1 and Th2 differentiation.

    MLN4924 Inhibits T-Cell Function. We further examined the effectsof blocking the NEDD8 pathway on T-cell activation using theNae1-specific inhibitor MLN4924 (8). CD4+ T cells were stim-ulated with anti-CD3 and anti-CD28 mAbs in the presence ofindicated concentrations of MLN4924 for 16 h. Consistent withthe shRNA results, MLN4924 suppressed the anti-CD3/CD28-induced proliferation of CD4+ T cells as assessed by EdU/7-AAD staining (Fig. 2A). The inhibitor also blocked T-cell cycleprogression from G0/G1 to S phase after activation, as well asup-regulation of the activation markers CD69 and CD25 in re-sponse to TCR stimulation (Fig. 2B). We next examined theeffect of MLN4924 on IL-2 production. Treatment with the in-hibitor markedly decreased IL-2 production in T cells (Fig. 2C).Quantitative RT-PCR analysis revealed less IL-2 mRNA inMLN4924-treated T cells (Fig. 2D). It should be noted thatMLN4924-treated CD4+ T cells did not show a substantial in-crease in cell death under low-concentration treatment, as mea-sured by 7-AAD staining (Fig. 2E), indicating that the inhibitoryeffect of MLN4924 on T-cell activation is not due to an increasein cell death. Together, the use of MLN4924 recapitulated thesiRNA knockdown results, supporting a potent positive functionof the NEDD8 pathway in T-cell activation.

    C

    020406080

    100120140160

    0 0.05 0.1

    IL-2

    (pg

    /ml)

    MLN4924 (µM)B

    CD69

    0 0.05 0.1MLN4924 (µM)

    CD25

    0

    20

    40

    60

    80

    100

    1200h

    6h

    16h

    D

    MLN4924 0 0.05 0.1 (µM)

    Rel

    ativ

    e m

    RN

    A

    IL-2

    E

    7-AAD

    0 0.05 0.1MLN4924 (µM)

    Cel

    l #

    A

    DNA content (7-AAD)

    EdU

    0 0.05 0.1MLN4924 (µM)

    23.1 14.6 4.1

    67.4 71.6 82.8

    Fig. 2. MLN4924 treatment blocks TCR-induced proliferation and activation. (A) Flow cytometry of the cell cycle status of CD4+ T cells stimulated with anti-CD3 and anti-CD28 for 16 h in the presence of the indicated concentrations of MLN4924 diluted in DMSO or equivalent volumes of DMSO alone. (B) CD4+ Tcells were analyzed for surface expression of CD69 (Upper) and CD25 (Lower) after stimulation with anti-CD3 and anti-CD28 in the presence of the indicatedconcentrations of MLN4924. (C) IL-2 production by CD4+ T cells stimulated with anti-CD3/CD28 mAbs in the absence or presence of MLN4924, measured byELISA. (D) Quantitative RT-PCR analysis of IL-2 mRNA. (E) Cell death of stimulated CD4+ T cells examined by 7-AAD staining and FACS analysis.

    626 | www.pnas.org/cgi/doi/10.1073/pnas.1213819110 Jin et al.

    Dow

    nloa

    ded

    by g

    uest

    on

    May

    30,

    202

    1

    http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1213819110/-/DCSupplemental/pnas.201213819SI.pdf?targetid=nameddest=SF3www.pnas.org/cgi/doi/10.1073/pnas.1213819110

  • NEDD8 Pathway Is Required for Erk Activation in T Cells. To un-derstand the molecular mechanism by which NEDD8 pathwayregulates CD4+ T-cell function, we analyzed several TCR-inducedsignaling events in Ubc12 knockdown CD4+ T cells. Knockdownof Ubc12 reduced the neddylation of cullin-1 (Fig. 3A), sug-gesting the efficient blockage of the neddylation pathway byUbc12 ablation. Activation of the Erk1/2 by TCR/CD28 stimu-lation was profoundly impaired in the Ubc12 knockdown CD4+

    T cells, whereas phosphorylation of p38 was slightly decreased(Fig. 3A). To confirm this result, we next pretreated CD4+ T cellsfor 16 h with MLN4924 before subjecting these cells to TCRstimulation with anti-CD3/CD28 mAbs. Pretreatment with0.1 μM MLN4924 reduced the activation of Erk, whereas the

    phosphorylation of PLC-γ1 was unchanged by treatment with theinhibitor (Fig. 3B). As expected, MLN4924 pretreatment effi-ciently blocked the neddylation of cullin-1 (Fig. 3B). Taken to-gether, these data indicate that the neddylation pathway isinvolved in TCR-stimulated Erk activation.Given that NEDD8 modification of cullin-1 has been dem-

    onstrated to be necessary for the ubiquitination of IκB-α andsubsequent activation of NF-κB (11, 12), we also examined IκB-αdegradation by TCR stimulation. Ubc12 knockdown T cells showedan only slight delay in IκB-α degradation in response to TCRstimulation compared with control T cells (Fig. 3A). We alsoexamined phorbol 12-myristate 13-acetate/ionomycin- or TNF-α–induced IκB-α degradation in the NEDD8 inhibitor-treated

    A 0 5 10 20 30 0 5 10 20 30 (min)

    Nedd8-cullin-1

    p-ERK

    p-p38

    Ubc12

    ERK

    ctl shUbc12

    p38

    cullin-1

    Grb2

    -CD3/CD28

    Nedd8-cullin-1

    p-ERK

    ERK

    cullin-1

    p-PLC -1

    MLN4924 0 0.05 0.1 (µM)-CD3/CD28 0 5 10 (min)

    Grb2

    0 5 10 0 5 10 B

    cullin-1 Nedd8-cullin-1

    Nedd8-ShcIgHp52Shc/p46Shc

    WCE IP

    Nedd8-Ubc12Ubc12

    MLN4924 - - + - - +

    C

    F

    IP: FlagIgG PTB

    Shc Ap66SH2CH1CH2 PTB

    PTB SH2CH1PTB

    PTB SH2CH1PTB

    p52

    p46

    D

    E HA-Nedd8: - + - + - + Flag-Shc: p66 p52 p46

    IP:FlagIB:HA

    IP:FlagIB:Flag

    IgHKGEPGRAADDGEGIVGAAMPDSGPLPLLQDMNKL

    HA-Nedd8: - + + - + + p52 K3R

    Nedd8-p52shc

    p52shc

    IP:FlagIB:HA

    WCEIB:HA

    Ubc12: - - + - - +

    Flag-Shc:

    Nedd8

    IP:FlagIB:Flag

    *

    Grb2

    Shc

    ZAP70

    p52Shc: - WT K3R -CD3/CD28 - - + - +

    WCE IP

    G

    - - + - + - WT K3R

    WB: Myc

    WB: Grb2

    WB: ZAP70

    IP: ZAP70IgG

    Grb2

    Shc

    ZAP70

    MLN4924 0 0 0.1 (µM)-CD3/CD28

    IP: ZAP70IgG

    WCE IP

    H - - + - + - - + - +

    0 0 0.1

    Fig. 3. The neddylation pathway is involved in Erk activation. (A) Immunoblot analysis of control (Left) and shUbc12 (Right) CD4+ T cells. The CD4+ T cellssorted from the chimeric mice were stimulated with anti-CD3 and anti-CD28 for the indicated time periods. Cell lysates were separated by SDS/PAGE andimmunoblotted with the indicated antibodies. (B) CD4+ T cells were preactivated with anti-CD3 in the presence of the indicated concentrations of MLN4924for 16 h and then restimulated with a combination of anti-CD3 and anti-CD28. (C) Jurkat E6.1 cells stably expressing FLAG-NEDD8 were treated with MLN4924for 12 h or left untreated before being collected. Cell lysates were boiled in 1% SDS-containing lysis buffer, then diluted to 0.1% SDS and immunoprecipitatedwith anti-FLAG–conjugated beads, followed by elution with triple FLAG peptide. The eluates were subjected to Western blot analysis using the indicatedantibodies. (D) The three isoforms of Shc. All isoforms contain an N-terminal PTB domain, a CH domain, and a C-terminal SH2 domain. (E) Jurkat cells weretransfected with FLAG-tagged p66, p52, or p46 with HA-tagged NEDD8. The samples were processed for immunoprecipitation as described in C. (F) Thepeptide sequence at the top is derived from MS analysis of p66Shc; an asterisk indicates the modified lysine residue. Jurkat cells were transfected withexpression vector encoding WT (wt) p52Shc or p52Shc lysine mutant (K3R). Where indicated, combinations of plasmids expressing HA-NEDD8 and/or Ubc12were cotransfected. (G) CD4+ T cells were stimulated with anti-CD3/CD28 antibodies and retrovirally transduced with Myc-tagged p52Shc WT-IRES-GFP (WT)or Myc-tagged p52Shc K3R-IRES-GFP (K3R). Whole-cell lysates were immunoprecipitated with anti-ZAP70 antibody and blotted with the indicated antibodies.(H) CD4+ T cells were treated with MLN4924 for 16 h or left untreated. After TCR stimulation, the cell lysates were subjected to coimmunoprecipitation withanti-ZAP70 antibody.

    Jin et al. PNAS | January 8, 2013 | vol. 110 | no. 2 | 627

    IMMUNOLO

    GY

    Dow

    nloa

    ded

    by g

    uest

    on

    May

    30,

    202

    1

  • T cells, and found that treatment of T cells with MLN4924 ledto retardation of the stimulus-induced IκB-α degradation, sug-gesting that this pharmacologic inhibitor indeed functions in theNF-κB activation process in T cells (Fig. S4). Ubc12 knockdowndid not lead to effective blockage of this pathway, however.

    Adaptor Protein Shc Is a Target for Neddylation. We asked whetherother NEDD8-modified proteins may be responsible for the ob-served Erk activation on TCR stimulation. In an effort to identifycurrently unknown NEDD8 substrates, we generated Jurkat E6.1cells stably expressing N-terminal 3× FLAG-tagged NEDD8. Thecells were lysed under strong denaturing conditions to blocknoncovalent protein–protein interactions, and FLAG-NEDD8–conjugated species were subjected to anti-FLAG immunoaffinitypurification. Proteomic analysis by MS identified the adaptorprotein Shc as a possible target (Fig. S5). We confirmed this ob-servation by an immunoprecipitation experiment. As a control, wetreated the stable cell lines with 1 μM MLN4924 for 12 h. Asexpected, we detected the NEDD8-modified forms of cullin-1 andUbc12 in the immunoprecipitates from the stable cells, but not inimmunoprecipitants from MLN4924-treated cells (Fig. 3C). Themolecular mass of immunoprecipitated proteins increased by ∼20kDa, consistent with a 3× FLAG-tagged NEDD8 conjugation.Western blot analysis with anti-Shc antibodies revealed a slower-migrating anti-Shc reactive band of ∼70 kDa. Treatment of cellswith MLN4924 caused the disappearance of this band in theimmunoprecipitated samples.Shc is expressed as three isoforms (p66, p52, and p46) encoded

    by the same genetic locus. All three isoforms have an SH2 do-main, a proline-rich CH1 domain, and a phospho-tyrosine binding(PTB) domain (Fig. 3D) (13, 14). Given that Jurkat E6.1 cells didnot express p66Shc at detectable levels, the slow-migrating bandcould represent an NEDD8-modified form of either p52Shc orp46Shc. To examine which isoforms of Shc can be modified withNEDD8, we generated expression vectors encoding FLAG-taggedShc isoforms. Jurkat cells were transfected with plasmids encodingFLAG-tagged p66Shc, p52Shc, or p46Shc isoforms along witha vector expressing HA-tagged NEDD8. Cells were lysed andimmunoprecipitated with anti-FLAG antibody, and then immu-noblotted with anti-HA or anti-FLAG antibody. Whereas p66Shc

    and p52Shc were conjugated to NEDD8, no conjugation was ob-served with p46Shc (Fig. 3E). Because the sole difference betweenp52Shc and p46Shc is the presence of an additional 45 amino acidsat the N terminus of p52Shc, this finding suggests that the first 45amino acids contain a potential NEDD8 conjugation site.To identify a lysine residue in p52Shc that acts as the site for

    neddylation, we performed MS analysis on NEDD8-conjugatedp66Shc immunopurified from cells expressing FLAG-p66Shc andHA-NEDD8. The analysis revealed that p66Shc was conjugatedwith NEDD8 at lysine 113, which is located at position 3 inp52Shc (Fig. 3F). To convincingly prove that the identified lysineresidue is a NEDD8 acceptor site for p52Shc, we constructeda mammalian expression vector carrying a mutation of the lysineresidue to an arginine residue (K3R). Plasmids encoding theK3R mutant were transfected into Jurkat cells in combinationwith HA-NEDD8. Here p52Shc (K3R) failed to become ned-dylated even in the presence of Ubc12 overexpression, suggest-ing that lysine 3 is the NEDD8 acceptor site of p52Shc (Fig. 3F).To further explore the potential mechanisms of Shc neddyla-

    tion on Erk activation, we examined the complex formation ofZAP70 with Shc and Grb2, and found that p52Shc interactedwith ZAP70 and Grb2 in TCR-stimulated CD4+ T cells, but thatthe Shc K3R mutation impaired the recruitment of Grb2 to theZAP70-Grb2 complex (Fig. 3G). Treatment of CD4+ T cells withMLN4924 also resulted in perturbed TCR-induced complexformation of Shc-Grb2 with ZAP70 (Fig. 3H). These findingssuggest that neddylation of Shc may facilitate the formation ofa ZAP70-Shc-Grb2 signaling complex and affect downstreamErk activation.

    NEDD8 Pathway Is Important for Airway Inflammation. We nextexamined whether the NEDD8 pathway in CD4+ T cells mayinfluence the development of airway inflammatory responses.We used an adoptive transfer model of experimental allergy (Fig.4A). GFP+ OT-II T cells were isolated from the mice trans-planted with control or Ubc12 shRNA-transduced bone marrowcells and then transferred i.v. into recipient B6 mice. At 1 d aftertransfer, recipient mice were immunized i.p. with OVA in alumadjuvant and then repeatedly challenged with OVA for 10 d afterimmunization. At 24 h after the last OVA challenge, the lungs

    A

    ctl shUbc12 0

    20

    40

    60

    80

    100

    0

    2

    4

    6

    8

    (pg

    /ml)

    ctl shUbc12

    IL-4 IL-5

    (pg

    /ml)

    DB shUbc12ctl

    H&E staining

    0

    10

    20

    30

    40

    50

    60

    Eos Mon Neu Lym

    # of

    Leu

    kocy

    tes(

    X10

    3 )

    C

    shUbc12ctl

    Fig. 4. Role pf the NEDD8 pathway in the development of allergic inflammation. (A) GFP+CD4+ T cells from OT-II control or OT-II Ubc12 knockdown chimericmice were adoptively transferred into C57BL/6J recipient mice, followed by immunization with OVA plus alum (n = 3). Starting at 10 d after immunization, themice were challenged intranasally with OVA protein for the next 4 d. The mice were examined at 24 h after the last challenge. (B) Lungs from control mice(Left) and shUbc12 knockdown chimeric mice (Right) were sectioned and stained with H&E to visualize inflammatory infiltrates. (C) Total numbers ofeosinophils (Eos), neutrophils (Neu), monocytes (Mon), and lymphocytes (Lym) were calculated by FACS analysis. (D) IL-4 (Left) and IL-5 (Right) concentrationsin BAL fluid were measured by ELISA at 24 h after the final OVA challenge. The results are representative of two repeated experiments.

    628 | www.pnas.org/cgi/doi/10.1073/pnas.1213819110 Jin et al.

    Dow

    nloa

    ded

    by g

    uest

    on

    May

    30,

    202

    1

    http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1213819110/-/DCSupplemental/pnas.201213819SI.pdf?targetid=nameddest=SF4http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1213819110/-/DCSupplemental/pnas.201213819SI.pdf?targetid=nameddest=SF5www.pnas.org/cgi/doi/10.1073/pnas.1213819110

  • were lavaged, and the bronchoalveolar lavage (BAL) fluid wasexamined. OVA sensitization and challenge resulted in anincreased number of leukocytes in BAL fluid in the micetransferred with control OT-II T cells; however, mice transferredwith Ubc12 knockdown T cells showed a substantially reducednumber of infiltrating inflammatory cells in the lungs (Fig. 4C).Histological analysis revealed prominent cellular infiltrationaround the bronchioles in control mice, but significantly moreattenuated cell infiltration in the mice transferred with Ubc12knockdown T cells (Fig. 4B). Consistent with a defective allergicresponse, the mice transferred with Ubc12 knockdown T cellsalso had significantly lower levels of the Th2 cytokines IL-4 andIL-5 in the BAL fluid compared with control mice (Fig. 4D).Taken together, these results suggest that the NEDD8 pathway isimportant for efficient Th2-mediated lung inflammation.The data presented here demonstrate that the NEDD8 system

    regulates various aspects of CD4+ T-cell function, includingproliferation, cytokine production, and Th cell differentiation.Inhibition of the NEDD8 pathway by shRNA-mediated knock-down of neddylation components or MLN4924 led to impairedproliferation and IL-2 production of CD4+ T cells. Interestingly,the neddylation pathway seemed to have a more dominant effecton Erk activation in TCR-stimulated CD4+ T cells. Using acombination of molecular and proteomics approaches, we haveidentified a target Shc for neddylation, suggesting the possibilitythat neddylation of p52Shc might affect the formation of ZAP70-Shc-Grb2 signaling complexes; however, more detailed studiesare needed to clarify the exact molecular mechanisms as a futuredirection of investigation. Importantly, we have shown that theNEDD8 pathway is critical for the development of allergic re-sponses, as demonstrated in adoptive transfer experiments. Re-cent preclinical trials of MLN4924 for various cancers haveyielded promising results for the manipulation of the neddylationpathway in treating human diseases (10). Our data on theknockdown of Ubc12 in attenuating airway inflammation suggestthat this pathway may represent a potentially important molec-ular target for therapeutic intervention of T-cell–mediated in-flammatory and autoimmune diseases.

    Materials and MethodsRetroviral Transduction and Bone Marrow Reconstitution. C57BL/6 mice(CD45.2), B6.SJL mice (CD45.1), and OT-II mice (OVA-specific TCR Tg mice)

    were obtained from Jackson Laboratories. All mice were housed underspecific pathogen-free conditions. To generate shRNA-expressing bonemarrow chimeric mice, Phoenix-Eco packaging cells were transfected with3 μg of LMP vector with 9 μl of TransIT-LT1 (Mirus). At 48 h, the culturesupernatant containing retrovirus was collected. Mature T-cell–depletedbone marrow cells from B6.SJL mice (CD45.1) were cultured for 24 h in 10 ng/mL IL-3, 10 ng/mL IL-6, and 100 ng/mL SCF (all from Peprotech) containingcomplete DMEM before initial retroviral infection. Mature T-cell–depletedbone marrow cells were infected with retrovirus along with 5 μg/mL poly-brene (Sigma-Aldrich) by spin inoculation (420 × g for 1 h). At 2 d after in-fection, retroviral transduced bone marrow cells were injected into lethallyirradiated (900 rad) C57BL/6 recipient mice. Recipient mice were killed at8 wk after reconstitution for further analysis. All animal experiment proto-cols were approved by the Institutional Animal Care and Use Committee ofthe La Jolla Institute for Allergy and Immunology.

    Adoptive Transfer of OT-II T Cells and Immunization. Bone marrow cells fromTCR transgenic OT-II mice were transduced with control or Ubc12 shRNA-expressing retroviruses, which were then transplanted into lethally irradiatedmice. After 8 wk, isolated CD4+ T cells from OT-II control or OT-II Ubc12knockdown chimeric mice were adoptively transferred into C57BL/6J re-cipient mice. At 1 d after transfer, the recipient mice were immunized i.p.with OVA (50 μg, Grade V; Sigma-Aldrich) emulsified in complete Freund’sadjuvant (BD Diagnostics) by s.c. injection. At 10 d after immunization, cellswere collected from spleen and lymph nodes and cultured with OVA323–339peptide (10 μg/mL; AnaSpec) for 8 h at 37 °C in the presence of GolgiStopprotein inhibitor (BD Biosciences).

    Antigen-Induced Airway Inflammation. GFP+CD4+ T cells were sorted fromOT-II control and OT-II Ubc12 knockdown chimeric mice, and 1 × 106 GFP+CD4+

    T cells were injected i.v. into C57BL/6 recipient mice. At 1 d after transfer, therecipient mice were sensitized by i.p. injection of 20 μg of OVA protein(Grade V; Sigma-Aldrich) adsorbed to 2 mg of aluminum hydroxide (alum)gel (Imject Alum; Pierce). At 10 d after immunization, the mice werechallenged intranasally with OVA protein for 4 consecutive days. Micewere killed at 24 h after the last aerosol challenge and assessed for lunginflammation. BAL fluid was collected for assessment of cytokine levelsand histological analysis.

    More detailed information on the experiments is provided in SI Materialsand Methods.

    ACKNOWLEDGMENTS. We thank J. Lopez and C. Elly for mouse breedingand K. Iwanami for technical help. This work was supported by NationalInstitutes of Health, National Institute of Allergy and Infectious DiseasesGrants R01 AI62969 and R01 AI78272. H.-s.J. was supported in part by NationalResearch Foundation of Korea Grant NRF-2009-352-C00098.

    1. Smith-Garvin JE, Koretzky GA, Jordan MS (2009) T cell activation. Annu Rev Immunol

    27:591–619.2. Liu YC, Penninger J, Karin M (2005) Immunity by ubiquitylation: A reversible process

    of modification. Nat Rev Immunol 5(12):941–952.3. Sun SC (2008) Deubiquitylation and regulation of the immune response. Nat Rev

    Immunol 8(7):501–511.4. Kerscher O, Felberbaum R, Hochstrasser M (2006) Modification of proteins by ubiq-

    uitin and ubiquitin-like proteins. Annu Rev Cell Dev Biol 22:159–180.5. Harper JW, Schulman BA (2006) Structural complexity in ubiquitin recognition. Cell

    124(6):1133–1136.6. Saha A, Deshaies RJ (2008) Multimodal activation of the ubiquitin ligase SCF by Nedd8

    conjugation. Mol Cell 32(1):21–31.7. Duda DM, et al. (2008) Structural insights into NEDD8 activation of cullin-RING ligases:

    Conformational control of conjugation. Cell 134(6):995–1006.8. Soucy TA, et al. (2009) An inhibitor of NEDD8-activating enzyme as a new approach to

    treat cancer. Nature 458(7239):732–736.

    9. Lin JJ, Milhollen MA, Smith PG, Narayanan U, Dutta A (2010) NEDD8-targeting drugMLN4924 elicits DNA rereplication by stabilizing Cdt1 in S phase, triggering check-point activation, apoptosis, and senescence in cancer cells. Cancer Res 70(24):10310–10320.

    10. Soucy TA, Dick LR, Smith PG, Milhollen MA, Brownell JE (2010) The NEDD8 conju-gation pathway and its relevance in cancer biology and therapy. Genes Cancer 1(7):708–716.

    11. Read MA, et al. (2000) Nedd8 modification of cul-1 activates SCF(beta(TrCP))-dependent ubiquitination of IkappaBalpha. Mol Cell Biol 20(7):2326–2333.

    12. Amir RE, Haecker H, Karin M, Ciechanover A (2004) Mechanism of processing of theNF-kappa B2 p100 precursor: Identification of the specific polyubiquitin chain-an-choring lysine residue and analysis of the role of NEDD8-modification on the SCF(beta-TrCP) ubiquitin ligase. Oncogene 23(14):2540–2547.

    13. Zhang L, Lorenz U, Ravichandran KS (2003) Role of Shc in T-cell development andfunction. Immunol Rev 191:183–195.

    14. Finetti F, Savino MT, Baldari CT (2009) Positive and negative regulation of antigenreceptor signaling by the Shc family of protein adapters. Immunol Rev 232(1):115–134.

    Jin et al. PNAS | January 8, 2013 | vol. 110 | no. 2 | 629

    IMMUNOLO

    GY

    Dow

    nloa

    ded

    by g

    uest

    on

    May

    30,

    202

    1

    http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1213819110/-/DCSupplemental/pnas.201213819SI.pdf?targetid=nameddest=STXThttp://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1213819110/-/DCSupplemental/pnas.201213819SI.pdf?targetid=nameddest=STXT