<<

p38α Environmental Stress To Control Innate Immune Responses via Mechanistic Target of Rapamycin

This information is current as Karl Katholnig, Christopher C. Kaltenecker, Hiroko of October 1, 2021. Hayakawa, Margit Rosner, Caroline Lassnig, Gerhard J. Zlabinger, Matthias Gaestel, Mathias Müller, Markus Hengstschläger, Walter H. Hörl, Jin Mo Park, Marcus D. Säemann and Thomas Weichhart J Immunol published online 11 January 2013 http://www.jimmunol.org/content/early/2013/01/11/jimmun Downloaded from ol.1202683 http://www.jimmunol.org/ Why The JI? Submit online.

• Rapid Reviews! 30 days* from submission to initial decision

• No Triage! Every submission reviewed by practicing scientists

• Fast Publication! 4 weeks from acceptance to publication

by guest on October 1, 2021 *average

Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2013 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Published January 11, 2013, doi:10.4049/jimmunol.1202683 The Journal of Immunology

p38a Senses Environmental Stress To Control Innate Immune Responses via Mechanistic Target of Rapamycin

Karl Katholnig,* Christopher C. Kaltenecker,* Hiroko Hayakawa,† Margit Rosner,‡ Caroline Lassnig,x Gerhard J. Zlabinger,{ Matthias Gaestel,‖ Mathias Mu¨ller,x Markus Hengstschla¨ger,‡ Walter H. Ho¨rl,* Jin Mo Park,† Marcus D. Sa¨emann,* and Thomas Weichhart*,‡

The MAPK p38a senses environmental stressors and orchestrates inflammatory and immunomodulatory reactions. However, the molecular mechanism how p38a controls immunomodulatory responses in myeloid cells remains elusive. We found that in monocytes and macrophages, p38a activated the mechanistic target of rapamycin (mTOR) pathway in vitro and in vivo. p38a signaling in myeloid immune cells promoted IL-10 but inhibited IL-12 expression via mTOR and blocked the differentiation of

proinflammatory CD4+ Th1 cells. Cellular stress induced p38a-mediated mTOR activation that was independent of PI3K but Downloaded from dependent on the MAPK-activated protein kinase 2 and on the inhibition of tuberous sclerosis 1 and 2, a negative regulatory complex of mTOR signaling. Remarkably, p38a and PI3K concurrently modulated mTOR to balance IL-12 and IL-10 expression. Our data link p38a to mTOR signaling in myeloid immune cells that is decisive for tuning the immune response in dependence on the environmental milieu. The Journal of Immunology, 2013, 190: 000–000. http://www.jimmunol.org/ ecognition of pathogen-associated molecular patterns and stress-activated kinases 1 and 2, MAPK-interacting serine/ by innate immune receptors triggers inflammatory and threonine-protein kinases 1 and 2, or MAPK-activated protein R immune responses involving several signaling mole- kinase (MK)2 and MK3 that mediates TNF-a production (9, cules including the MAPKs (1, 2). The MAPK p38a (also known 10). as MAPK14) is one of four homologs of mammalian p38 and is The finding that inhibiting p38 blocks LPS-induced proin- essential in innate and adaptive immune signaling cascades (3). flammatory cytokine production (7) initiated the development of p38a is activated by diverse stimuli including TLR ligands, a wide range of p38 inhibitors for treatment of chronic inflam- cytokines, and physicochemical stress signals such as UV irradi- matory diseases such as rheumatoid arthritis, , or Crohn’s ation, heat or osmotic shock, arsenite, or anisomycin (4). p38a disease (11). SB203580 is a competitive inhibitor of p38a and by guest on October 1, 2021 is ubiquitously expressed in most cell types and regulates di- p38b by blocking ATP binding to the kinase, whereas BIRB796 verse functions such as cell proliferation, differentiation, apo- is an allosteric inhibitor of p38a, p38b, and p38g (12, 13). Re- ptosis, tissue repair, tumorigenesis, or inflammation (4, 5). For markably, so far, p38 inhibitors failed in clinical trials because example, p38a was previously identified as activator of the pro- of adverse and inflammatory effects such as liver toxicity or skin inflammatory cytokines IL-1b, IL-6, TNF-a, or cyclooxygenase 2 rashes (11). (6–8). Several kinases are activated by p38a such as mitogen- Recently, a more complex role of p38a has been reported (14– 16). Expression of p38a in myeloid cells limits inflammation in an *Clinical Division of Nephrology and Dialysis, Department of Internal Medicine III, UV-induced irradiation model (14). These immunomodulatory Medical University of Vienna, 1090 Vienna, Austria; †Cutaneous Biology Research effects of p38a may be mediated by the induction of the anti- Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129; ‡Institute of Medical Genetics, Medical University of Vienna, 1090 inflammatory cytokine IL-10 (14, 15) and the inhibition of pro- Vienna, Austria; xInstitute of Animal Breeding and Genetics and Biomodels Austria, inflammatory IL-12 (14, 16). However, the downstream pathway University of Veterinary Medicine Vienna, 1210 Vienna, Austria; {Institute of Im- ‖ that controls coordinated IL-10 and IL-12 expression by p38a has munology, Medical University of Vienna, 1090 Vienna, Austria; and Department of Physiological Chemistry, Hannover Medical School, 30625 Hannover, Germany remained elusive. Received for publication September 25, 2012. Accepted for publication December The classical insulin signaling pathway consisting of PI3K, 11, 2012. Akt, and mechanistic target of rapamycin (mTOR) complex 1 This work was supported by the Else-Kro¨ner Fresensius Stiftung (to T.W. and M.D.S.), (mTORC1) has recently emerged as key regulator of innate im- the Austrian Science Foundation Fonds zur Fo¨rderung der Wissenschaftlichen mune cell homeostasis (17–20). Stimulation of innate immune Forschung (SFB F28 to C.L. and M.M.), the Austrian Federal Ministry for Science and Research (GEN-AU III Austromouse to C.L. and M.M.), the National Institutes cells by TLR ligands activates the mTOR pathway, and it is, in of Health (Grant AI074957 to J.M.P.), and the Deutsche Forschungsgemeinschaft fact, a major pathway activated in LPS-stimulated macrophages (to M.G.). based on phosphoproteomics (21). The function of PI3K–Akt– Address correspondence and reprint requests to Dr. Thomas Weichhart, Institute of mTOR is cell type specific, but it has been shown that inhibition Medical Genetics, Medical University of Vienna, Wa¨hringer Gu¨rtel 18-20, 1090 Vienna, Austria. E-mail address: [email protected] of PI3K by wortmannin or mTOR by rapamycin in myeloid Abbreviations used in this article: BMDM, marrow–derived macrophage; DC, cells, such as human monocytes, macrophages, or myeloid den- dendritic cell; MEF, mouse embryonic fibroblast; MK, MAPK-activated protein ki- dritic cells (DCs), enhances IL-12 production but blocks the nase; mTOR, mechanistic target of rapamycin; mTORC1, mTOR complex 1; SAC, release of IL-10 in vitro and in vivo (22–28). Tuberous sclerosis Staphylococcus aureus; TSC, tuberous sclerosis; WT, wild-type. (TSC)2 is a tumor suppressor that is phosphorylated and inac- Copyright Ó 2013 by The American Association of Immunologists, Inc. 0022-1767/13/$16.00 tivated by the protein kinase Akt, which itself is activated by

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1202683 2 p38a REGULATES INNATE IMMUNITY VIA TSC/mTOR

PI3K (29). TSC2 forms a heterodimeric complex with TSC1 and Materials and Methods negatively regulates mTOR (29). Conversely, knockdown of Reagents TSC2 in human monocytes or macrophages enhances IL-10 LPS (Escherichia coli 0111:B4), wortmannin, anisomycin, and rapamycin but inhibits IL-12 production (24, 30). In line, genetic inactiva- were from Sigma. Staphylococcus aureus (SAC; PANSORBIN) and + tion of mTORC1 reduces IL-10 production in intestinal CD11c SD169 were from Calbiochem. BIRB796 was a kind gift of Sir Philip CD11b+ DCs (31). In contrast, TSC1-deficient macrophages show Cohen or purchased from Axon Medchem. SB203580 was from Tocris elevated production of TNF-a and IL-12p40 (32). Despite these Bioscience and IFN-g from R&D Systems. Heat-killed cells of Listeria observations, the precise regulatory units and upstream pathways monocytogenes (L.m.) were prepared by incubating the viable log-phase bacterial suspension at 70˚C for 1 h. For UV exposure, cell culture plates controlling mTOR-dependent cytokine production are still un- were placed on a 20 3 20 UV-transilluminator (MWG Biotech) and ac- clear. tivated with UV light for 10 s, 30 s, or 1 min. An outstanding question is how myeloid immune cells adapt and coordinate their immune response to an infectious trigger Cell culture toward the status of the environmental milieu, for example, how + Human PBMCs and peripheral human myeloid DCs were isolated as de- to avoid detrimental tissue-destructive CD4 Th1 responses scribed previously (24). Monocytes were isolated from PBMCs by MACS under conditions of tissue repair. Moreover, it is imperative to using CD14 Microbeads (Miltenyi Biotec). RPMI 1640 supplemented with explore the molecular signaling pathways that regulate p38a- 2mML-glutamine, 100 mg/ml streptomycin, 100 U/ml penicillin (all from mediated immune responses for a deeper understanding of the Life Technologies), and 10% FCS (Hyclone) was used as culture medium. Mouse embryonic fibroblasts (MEFs) were cultured in DMEM containing effects of p38 inhibitors for human health and disease. There- 4.5 g/L glucose, 2 mM L-glutamine, 100 mg/ml streptomycin, 100 U/ml fore, we tested whether p38a is connected to PI3K–TSC2–mTOR penicillin, and 10% FCS. Tsc2+/+ p532/2 and Tsc22/2 p532/2 as well as Downloaded from signaling to regulate innate inflammatory responses. We found Tsc1+/+ and Tsc12/2 MEFs were described previously (33, 34). p85a2/2 2 2 that TLR ligands or environmental stress activates the TSC2- p85b / MEFs were a kind gift of Lewis Cantley. p38afl/fl and p38aDM mTOR pathway via p38a and MK2 to regulate the balance of IL- mice were described previously (14). Bone marrow–derived macrophages (BMDMs) from mice were isolated and grown as described previously (35) 12 and IL-10. Importantly, p38a acts in parallel to PI3K to control and were replated 1 d before stimulation in full medium containing 2% the IL-12/IL-10 equilibrium in response to the environmental FCS. Mk22/2 immortalized murine macrophages stably reconstituted with milieu. MK2 or MK2K79R were described previously (36). http://www.jimmunol.org/

FIGURE 1. p38a modulates IL-12 and IL-10 in mice and humans. (A)p38afl/fl and p38aDM BMDMs were stimulated 2 with medium ( ), LPS, SAC, or L.m. for by guest on October 1, 2021 24 h. IL-12p40, IL-12p70, and IL-10 in the supernatants were determined by Lumi- nex. Data are shown as means 6 SEM for five mice. (B) Serum levels of IL-12p40, IL-12p70, and IL-10 were determined in p38afl/fl and p38aDM mice injected with LPSfor4h(means6 SEM for three mice). (C–F) Human monocytes were treated with medium (2), BIRB796 (BIRB; 100 or 200 nM), SB203580 (SB; 200 nM or 2 mM), or rapamycin (Rap; 100 nM) and then stimulated with (C)LPS,(D)SAC,(E) L.m., or (F) LPS and IFN-g for 22 h. Cyto- kines in cell-free supernatants were mea- sured by Luminex (means 6 SEM of at least three donors). (G and H)Human monocytes were treated as indicated, washed, and added to allogeneic T cells for 1 wk. (G) IFN-g of cell-free superna- tants was determined by Luminex. Data represent means 6 SEM for three inde- pendent experiments. (H) Primed T cells were activated for 5 h with PMA/ion- omycin in the presence of brefeldin A. Intracellular cytokine staining for IL-4 and IFN-g in CD-4 T cells is illustrated. One representative experiment out of three is shown. *p , 0.05 compared with the re- spective controls. The Journal of Immunology 3

Measurement of cytokine production blotting were done as described previously (24). Abs were p-p70S6K (Thr389), p70S6K, p-4E-BP1 (Thr37/46), p-p38 (Thr180/Tyr182), p-S6 Cells were pretreated for 90 min with the indicated concentrations of (Ser 240/244), p-Akt (Ser473), p-MK2 (Thr334), GAPDH, S6-ribosomal SB203580, BIRB796, rapamycin, or wortmannin and then stimulated with 7 protein, p38 MAPK, p38a MAPK, p38b MAPK, p38d MAPK, Tuberin/ 100 ng/ml LPS (+30 ng/ml IFN-g as indicated), 75 mg/ml SAC, or 10 L.m. TSC2 (all Cell Signaling Technology), p-Erk (Tyr204), IkBa, and p38 in 48-well plates. Cell-free supernatants were collected after 22–24 h as (Santa Cruz Biotechnology). indicated. Human and murine cytokines were determined by the Luminex bead system with beads from R&D Systems and Affymetrix, and read on Quantitative RT-PCR a Luminex 100 reader. RNA from human monocytes or murine immortalized macrophages was LPS injection extracted in TRIzol (Invitrogen). cDNA was generated by Superscript II (Invitrogen). mRNA levels were determined by TaqMan Gene Expression p38aDM and p38afl/fl mice were housed and maintained at the Massachusetts Assays (Applied Biosystems) on a StepOnePlus Real-Time PCR System General Hospital and Harvard Medical School. Mice were injected i.p. with and normalized to ubiquitin. 30 mg/mouse LPS. After 4 h, serum samples were taken and spleens were isolated. Cytokine levels in the sera were measured by Luminex. Homoge- Transfection of MEFs nization of mouse tissue was performed by using the Precellys-ceramic kit Tsc22/2 MEFs in six-well plates at 20–40% confluency were transfected in 2.8 mm and the Precellys 24 tissue homogenizer (both from peQLab). DMEM without with 1 mg pcDNA3-HA-TSC2 wild-type (WT), T cell differentiation pcDNA3-HA-TSC2 S1210A, or empty vector with Lipofectamine 2000 Reagent (Invitrogen) for 36 h and afterward starved for 12 h in DMEM Monocytes were incubated with medium, 200 nM BIRB796, 2 mM SB203580, without antibiotics and FCS before stimulation. or 100 nM rapamycin for 90 min and stimulated with 100 ng/ml LPS for 24 h. Immunofluorescence microscopy The cells were then washed with PBS and incubated with allogeneic T lymphocytes at a ratio of 1:1 in 24-well plates in RPMI complete medium. Cells were applied to eight-well Permanox chamber slides (Lab-Tek Chamber Downloaded from After 1 wk, IFN-g production was determined in cell-free supernatants by Slide System), fixed with 4% paraformaldehyde, quenched with 100 mM Luminex. The primed cells were further activated for 5 h with 50 ng/ml PMA glycine, permeabilized with methanol, blocked with 1% BSA, and stained and 200 ng/ml ionomycin (both from Sigma) in the presence of 10 mg/ml with p-S6 Ab, p-MK2 Ab, or isotype control overnight at 4˚C. Cells were brefeldin A (Sigma) for the last 3 h. Afterward, cells were stained with FITC- stained with Alexa Fluor 488–labeled goat anti-rabbit IgG (Invitrogen) labeled anti–IFN-g, PE-labeled anti–IL-4, and allophycocyanin-labeled anti- followed by nuclear tracking using 0.1 mg/ml Hoechst-33342 (Invitrogen) CD4 (all BD Bioscience) and analyzed by flow cytometry. and mounted in Vectashield mounting medium.

Analysis of signal transduction events Statistics http://www.jimmunol.org/ Monocytes, BMDMs, or 70% confluent MEFs starved overnight were Results are expressed as means 6 SEM. Student t test was used to detect treated and stimulated as indicated. Extract preparation and Western statistical significance. by guest on October 1, 2021

FIGURE 2. p38 activation stimulates mTOR signaling. (A–C) Human monocytes were incu- bated with medium (2), BIRB796 (200 nM), SB203580 (2 mM), SD169 (200 nM), or rapa- mycin (Rap; 100 nM) and stimulated with (A) LPS (100 ng/ml), (B) anisomycin (50 ng/ml), or (C) UV (30 s) for the indicated times. Cell lysates were analyzed by immunoblotting. Representa- tives of three independent experiments are shown. (D and E) Murine macrophages were stimulated with (D) LPS or (E) SAC in the presence or ab- sence of anisomycin (50 ng/ml) for 1 h. IL-12p40 and IL-10 mRNA levels were measured by RT- PCR. Levels were normalized to ubiquitin and are shown relative to the (D)LPS-or(E)SAC-stim- ulated samples, which were set to 1 (means 6 SEM; n =3).*p , 0.05. (F)Monocyteswere incubated with LPS (10 ng/ml) or anisomycin (10 ng/ml) for 30 min as indicated. Cell lysates were analyzed by immunoblotting. 4 p38a REGULATES INNATE IMMUNITY VIA TSC/mTOR

Results increased IL-10 mRNA levels in LPS- or SAC-stimulated mac- p38a modulates IL-12 and IL-10 production in mice and rophages (Fig. 2D, 2E). Remarkably, hyperactivation of p38 with human anisomycin further increased mTOR activity in LPS-activated monocytes (Fig. 2F). These results indicate that p38 activates To evaluate the role of p38a in the myeloid , we the mTOR pathway and thereby regulates the expression of IL-12 generated BMDMs from mice with a deletion of p38a in cells and IL-10. expressing the lysozyme M gene (p38aDM) and stimulated these cells with LPS, SAC, or heat-killed L.m. (Fig. 1A). Deficiency of p38a deletion blocks mTOR activation in vitro and in vivo via p38a enhanced IL-12p40 and IL-12p70 expression, whereas the MK2 anti-inflammatory cytokine IL-10 was blocked compared with To extend our results genetically, we examined the mTOR signal- fl/fl controls carrying homozygously the floxed p38a gene (p38a ; ing pathway in p38aDM BMDMs. Deletion of p38a in macro- Fig. 1A). Other cytokines such as IL-1b, IL-23, or TNF-a were phages abolished MK2 activation and also strongly diminished the not significantly altered (data not shown). Injection of LPS into phosphorylation of p70S6K, S6, and 4EBP1 after stimulation with p38aDM mice similarly deviated the production of IL-12 and IL-10 in vivo (Fig. 1B). Next, we characterized the precise function of p38a, which is the most highly expressed p38 isoform in human monocytes. We found that inhibition of p38 with dif- ferent concentrations of SB203580 or BIRB796, as well as with the mTOR inhibitor rapamycin, strongly increased the production of IL-12p40 and IL-12p70 but blocked secretion of IL-10 after Downloaded from stimulation with LPS, SAC, or L.m. (Fig. 1C–F). Notably, SB203580 did not augment IL-12p40 production in L.m.-stimu- lated monocytes. Enhanced IL-12p40 but reduced IL-10 expression was also observed in SB203580- or BIRB796-treated peripheral human myeloid DCs stimulated with LPS, SAC, or L.m. (data not shown). http://www.jimmunol.org/ The surface expression of the costimulatory molecule CD86, important for T cell activation and priming, was enhanced upon p38 blockade in human monocytes after stimulation with LPS, SAC, or L.m. (data not shown). In line, we found that inhibition of p38 or mTOR in monocytes stimulated with LPS strongly en- hanced the production of IFN-g (Fig. 1G) and the differentiation of CD4+ Th1 cells (Fig. 1H) in an allogeneic T cell activation model. In summary, these data show that p38a differentially regulates the expression of IL-12 and IL-10 in human monocytes by guest on October 1, 2021 and DCs, as well as in BMDMs in vitro and in vivo in response to microbial insult. p38 activation stimulates mTOR signaling The concurrent regulation of IL-12/IL-10 by p38 and mTOR inhibitors indicated that these molecules might be connected. Therefore, we explored whether inhibition of p38 or mTOR might mutually influence the other kinase. Rapamycin did not modulate the phosphorylation of p38 or its downstream kinase MK2 after stimulation of human monocytes with LPS but blocked the phosphorylation of the mTOR substrates p70S6K and 4EBP1 (Fig. 2A). In contrast, inhibition of p38 with either SB203580 or BIRB796 blocked MK2 activation as expected, but also decreased the phosphorylation of 4EBP1 and p70S6K, as well as S6, sug- A fl/fl DM gesting that p38 activates mTOR signaling. Total levels of the FIGURE 3. p38a activates mTOR via MK2. ( ) p38a and p38a BMDMs were stimulated with medium (2), LPS (100 ng/ml), anisomycin investigated proteins were not altered by treatment with the in- 7 (100 ng/ml), UV (1 min), or L.m. (10 cells). Cell lysates were analyzed hibitors (data not shown). Interestingly, phosphorylation of Akt by immunoblotting. Representatives of three independent experiments at Ser473 was blocked, whereas activation of Erk was enhanced are shown. (B)p38afl/fl and p38aDM mice were injected with LPS for with BIRB796 and SB203580 (Fig. 2A, 2C). However, inhibition 4 h. Isolated and homogenized spleens were analyzed by immunoblot- of Erk did not inhibit mTOR and did not modulate the production ting. Results for two mice per genotype are shown. Similar results were 2 2 of IL-12 or IL-10 (data not shown). Degradation of IkB-a was obtained in an independent experiment (data not shown). (C) MK2 / K79R not influenced by either p38 or mTOR (Fig. 2A). Activation of macrophages reconstituted with either MK2 or WT MK2 were stim- p38 by the two environmental stress signals anisomycin or UV ulated with medium, LPS (100 ng/ml), or anisomycin (100 ng/ml) for 1 h, also activated mTOR signaling in human monocytes in a p38- and whole-cell lysates were analyzed by immunoblotting. Representatives of three independent experiments are shown. (D and E) MK2 WT and dependent manner (Fig. 2B, 2C). SD169, another reported in- K79R MK2 macrophages were stimulated with medium (2), LPS (100 ng/ hibitor of p38a, was without effect in monocytes (Fig. 2A, 2C). ml), or SAC (75 mg/ml) for 2 h. mRNA levels of IL-12p40 and IL-10 were Next, we tested whether hyperactivation of p38 with anisomycin measured by RT-PCR. Levels were normalized to ubiquitin and are shown in the presence of LPS or SAC could influence cytokine ex- relative to the (D) LPS- or (E) SAC-treated MK2 WT samples, which were pression. Indeed, anisomycin reduced IL-12p40 mRNA levels but set to 1 (means 6 SEM; n = 3). *p , 0.05. The Journal of Immunology 5

LPS, UV, anisomycin, or L.m. (Fig. 3A and data not shown). Erk and mTOR signaling in Tsc1+/+ cells, as well as in Tsc2+/+ cells, activation was observed only after LPS or L.m. treatment and not and inhibition of p38 by either SB203580 or BIRB796 strongly modified in p38aDM BMDMs (Fig. 3A). Moreover, activation of reduced mTOR activation as shown by diminished phosphoryla- p70S6K and 4EBP1 was blocked in spleens from p38aDM, but not tion of p70S6K or S6 (Fig. 4A–D). Strikingly, inhibition of p38 p38afl/fl, mice that were challenged with LPS (Fig. 3B), demon- did not block mTOR signaling in cells deficient of TSC1 or TSC2 strating that p38a activates mTOR signaling in vitro and in vivo. (Fig. 4A–D). Rapamycin, which acts downstream of TSC1/TSC2, Next we analyzed whether MK2 may mediate the effect of p38a still inhibited mTOR in these cells (Fig. 4A–D). These results on mTOR signaling. Therefore, we used macrophages expressing suggest that the TSC1/TSC2 complex is a critical signaling nodule a catalytic-dead mutant of MK2 (K79R) or its WT control. Strik- that senses p38 activity to regulate mTOR activation. Interestingly, ingly, activation of mTOR after stimulation with LPS or aniso- serum, a well-known mTOR activator, did not stimulate p38 ac- mycin was severely compromised in the K79R mutant compared tivation, and inhibition of p38 did not influence mTOR activation with WT MK2 (Fig. 3C). Moreover, the K79R macrophages induced by serum (Fig. 4E). Previously, it has been shown that showed absent IL-10 expression after LPS or SAC stimulation, MK2 phosphorylates Ser1210 in TSC2 (37). However, it remained whereas IL-12p40 was strongly increased (Fig. 3D, 3E). These to be verified whether p38 activation regulates TSC2 activity via data indicate that the kinase activity of MK2 transmits the p38a Ser1210 phosphorylation by MK2. To explore this possibility, we signal to stimulate mTOR signaling and to regulate the production transfected Tsc22/2 cells with plasmids encoding either WT of IL-12 and IL-10. TSC2 or a S1210A TSC2 mutant and observed that WT, but not mutant, TSC2 restored the ability of anisomycin to activate p38 signals to mTOR via TSC1/TSC2 to regulate IL-12 and p70S6K (Fig. 4F). Moreover, BIRB796 inhibited p70S6K acti- IL-10 vation upon p38 stimulation in cells transfected with WT, but not Downloaded from TSC1 and TSC2 act as dimer to inhibit activation of mTORC1 (29). mutant, TSC2 (Fig. 4F). Overall, these results demonstrate that To investigate whether p38-MK2 may mediate mTORC1 activa- activation of p38 inhibits the TSC1/TSC2 complex, allowing tion via TSC1/TSC2, we made use of MEFs deficient in either mTOR activation potentially via MK2-dependent phosphorylation TSC1 or TSC2. Treatment with anisomycin or UV-stimulated p38 of TSC2 at Ser1210. http://www.jimmunol.org/ by guest on October 1, 2021

FIGURE 4. p38 signals to mTOR via TSC1/TSC2. (A and B) Tsc1+/+ and Tsc12/2 MEFs or (C–E) Tsc2+/+ and Tsc22/2 MEFs were starved overnight and treated with me- dium (2), BIRB796 (200 nM), SB203580 (2 mM), or rapamycin (Rap; 100 nM) for 90 min. Afterward, MEFs were stimulated with (A, C) anisomycin (100 ng/ml), (B, D)UV(1 min), or (E) 10% serum for 60 min. Whole- cell lysates were analyzed by immunoblot- ting. (F) Tsc22/2 MEFs were transfected with empty vector (e.v.), HA-tagged WT TSC2, or HA-tagged TSC2 S1210A and then treated with medium, BIRB796, and anisomycin as indicated. Immunoblotting was performed with the indicated Abs. Long exposure re- vealed the phosphorylation of the p85S6K (p85) isoform in addition to the p70S6K (p70) isoform. Densitometric analysis was performed on the intensity of p-p70S6K. One representative of three different experiments is shown. NB, Nonspecific bands. 6 p38a REGULATES INNATE IMMUNITY VIA TSC/mTOR p38a and PI3K independently regulate mTOR p38 and PI3K coordinately control expression of IL-12 and Classical activation of mTOR signaling by growth factors or IL-10 TLR ligands is dependent on PI3K (17, 38). Our results so far Finally, we wanted to elucidate the relative contribution of p38a established that p38a activates mTOR via MK2 involving an and PI3K for the production of IL-12 and IL-10. BMDMs showed Ser1210-dependent regulation of TSC2 to modulate IL-12/IL-10 maximum production of IL-12p70 or IL-12p40 when PI3K was signaling. To further delineate the relative requirements of PI3K inhibited regardless of whether p38a was present (Fig. 6A and and p38a for stimulating mTOR, we inhibited p38 in the presence data not shown). This indicates that PI3K activation is dominant or absence of wortmannin, a specific covalent PI3K inhibitor. after LPS stimulation, and that p38a modulates IL-12p40 within Treatment with either wortmannin or BIRB796 considerably di- this context. In contrast, IL-10 production seems to more thor- minished phosphorylation of S6 in human LPS- or anisomycin- oughly depend on p38a as deletion of p38a already diminished activated monocytes (Fig. 5A, 5B). The combination of both in- IL-10 production of BMDMs to a level that could not be further hibitors led to a near-complete abolishment of S6 phosphorylation inhibited by PI3K inhibition (Fig. 6A). Similar results were ob- (Fig. 5A, 5B). Likewise, mTOR was blocked more efficiently tained in human monocytes (Fig. 6B). In these cells, p38 was also in BMDMs from p38aDM than from p38afl/fl mice after wort- dominant for IL-10 production, whereas PI3K and p38 both reg- mannin treatment (Fig. 5C, 5D). Immunofluorescence analysis ulated IL-12p40 (Fig. 6B). In conclusion, these results suggest that confirmed that wortmannin inhibited the phosphorylation of S6 PI3K and p38a coordinately modulate mTOR signaling to regu- more completely in p38aDM than in p38afl/fl BMDMs (Fig. 5E), late the expression of IL-12 and IL-10 in myeloid immune cells. suggesting that PI3K and p38 cooperatively stimulate mTOR

signaling. Next, we analyzed MEFs deficient in p85a/p85b, the Discussion Downloaded from regulatory subunits of PI3K, to assess the importance of p38 Monocytes, macrophages, and DCs are emerging therapeutic tar- versus PI3K for maximum activation of mTOR (Fig. 5F). Phos- gets in cardiovascular, malignant, and autoimmune disorders. Ad- phorylation of p70S6K and 4E-BP1 induced by LPS, anisomycin, dressing gaps in knowledge about the function of these cells in or UV exposure was severely reduced but still detectable in p852/2 response to environmental stimuli is required to understand the MEFs compared with their WT counterparts (Fig. 5F). However, in vivo responses to therapies that target these cells. Stringent concurrent inhibition of p38 by BIRB796 in p852/2 MEFs further control of MAPK signaling is critical for balancing proinflam- http://www.jimmunol.org/ inhibited activation of mTOR (Fig. 5F). Together, these results matory versus anti-inflammatory signaling to enable efficient strongly suggest that full activation of mTOR in myeloid immune pathogen killing but also to limit detrimental tissue pathology. In cells is dependent on p38 and PI3K. that regard, p38 is one of the most studied targets for anti- by guest on October 1, 2021

FIGURE 5. p38a and PI3K independently regulate mTOR. (A and B) Human monocytes were incubated with medium (2), BIRB796 (200 nM), and/or wortmannin (WM; 100 nM) for 90 min and stimulated with (A) LPS (100 ng/ml) or (B) anisomycin (100 ng/ml). Cell lysates were analyzed by immunoblotting. (C– E) p38afl/fl and p38aDM BMDMs were treated with medium (2), BIRB796 (200 nM), SB203580 (2 mM), and/or wortmannin (WM; 100 nM) as indicated and stimulated with (C, E) LPS (100 ng/ml) or (D) anisomycin (100 ng/ ml) for 1 h. (C, D) Cell lysates were analyzed by immunoblotting. (E) Phosphorylation of S6 and MK2 was analyzed by immunofluores- cence; original magnification 340. One repre- sentative experiment of three is shown. (F) p85+/+ and p852/2 MEFs were treated with medium (2) or BIRB796 (200 nM) followed by stimulation with LPS (100 ng/ml), aniso- mycin (100 ng/ml), or UV (10 s) for 60 min. Cell lysates were analyzed by immunoblotting. The Journal of Immunology 7

FIGURE 6. p38a and PI3K coordinately regu- late the IL-12/IL-10 balance. (A) p38afl/fl and p38aDM BMDMs were treated with wortmannin (WM; 100 nM) and stimulated with medium (2), SAC (75 mg/ml), or LPS (100 ng/ml) for 24 h. Cytokine levels in cell-free supernatants were determined by Luminex (means 6 SEM; n = 5). (B) Human monocytes were treated with wort- mannin (WM; 100 nM) or BIRB796 (200 nM) as indicated and stimulated with LPS (100 ng/ml) for 24 h. IL-12p40 and IL-10 were determined in the supernatants by Luminex (means 6 SEM;

n = 3). (C) Model of mTOR-regulated produc- Downloaded from tion of IL-12/IL-10 via PI3K and p38 on the level of TSC1/TSC2. *p , 0.05; n.s., Not sig- nificant. http://www.jimmunol.org/

inflammatory therapy. This kinase directly or indirectly regulates tivation to precisely allow the expression of proinflammatory and many transcription factors and, therefore, participates in the gene anti-inflammatory cytokines in response to environmental stress. induction of cytokines and other inflammatory molecules. p38 is We are unaware of a report demonstrating that two stimuli addi- by guest on October 1, 2021 also important in the posttranscriptional regulation of gene ex- tively regulate the activation of mTOR via the TSC complex. We pression during inflammation (39). Animal studies have shown suggest that p38a-mediated mTOR activation in addition to the that p38 inhibitors are efficacious in several disease models, in- PI3K pathway represents a tuning mechanism to regulate im- cluding inflammation, arthritis and other diseases, septic munomodulatory cytokines to adapt the immune response to the shock, and myocardial injury (11). However, translation into the environmental milieu. This is supported by the observation that clinic has been difficult either because of lack of efficiency or the hyperactivation of p38a by anisomycin can modulate IL-12 and appearance of adverse effects including inflammation such as skin IL-10 expression on top of a TLR signal (Fig. 2D). The p38/MK2 rashes. axis is required after excessive tissue damage to induce tissue p38a is activated in diverse cell types by a wide array of stress repair (41, 42). In such situations, p38a may promote mTOR stimuli including genotoxic agents, pathogen-associated molecu- activation in resident and recruited macrophages to reduce IL-12 lar patterns, proinflammatory cytokines, heat or osmotic shock, and augment IL-10 production that limits the generation of a partial pressure, or chemical insults (arsenite and aniso- proinflammatory CD4+ Th1 response that would further exag- mycin) (40). The simple view of an entirely proinflammatory ki- gerate tissue damage (43). nase promoting the expression of TNF-a and IL-1b shifted to A link from p38b to mTOR has been described in Drosophila a more complex role in recent years demonstrating that p38a that occurs via a TSC2-independent mechanism (44). In line, controls IL-12 and IL-10 expression (14–16). However, the down- p38b was recently shown to phosphorylate the essential mTORC1 stream pathway that regulates these immunomodulatory cytokines binding protein Raptor and to participate in arsenite-induced remained unknown. mTOR activation in fibroblasts (45). In contrast, in the same In this article, we have demonstrated that p38a uses the TSC2/ cell, p38b can also inhibit mTOR upon energy starvation via mTOR signaling pathway to control the balance of IL-12 and IL- phosphorylation and inactivation of Ras homolog enriched in 10. Our data suggest that TLR ligands or stress stimuli lead to an brain (Rheb), a key component of the mTORC1 pathway (46). We activation of p38a that, in turn, activates its downstream kinase now show that p38a via MK2 promotes mTOR activation de- MK2. The kinase activity of MK2 most likely phosphorylates pendent on the TSC1/TSC2 complex in myeloid immune cells. Ser1210 of TSC2, leading to inactivation of the TSC1/TSC2 Indeed, MK2 was shown to phosphorylate TSC2 at Ser1210 in complex and, in turn, activation of the mTOR pathway (Fig. fibroblasts (37). In addition, MK2 was also described to phos- 6C). Activation of mTOR then promotes IL-10 production, phorylate Akt at Ser473 in neutrophils (47), in line with the inhi- whereas reducing IL-12 expression. Our work indicates that p38a- bition of Akt Ser473 by the p38 inhibitors in our cells (Figs. 2A, 6). mediated mTOR activation occurs in parallel to the well-known We have previously shown that mTOR regulates NF-kB and PI3K pathway that activates mTOR in response to TLR signals STAT3 signaling (24). Interestingly, p38a and MK2 also are re- (17, 19). Hence, both pathways concurrently control mTOR ac- quired for STAT3 activation and IL-10 production (14, 36). These 8 p38a REGULATES INNATE IMMUNITY VIA TSC/mTOR effects are likely to be indirectly mediated by p38 and mTOR, and 16. Yang, Z., X. Zhang, P. A. Darrah, and D. M. Mosser. 2010. The regulation of Th1 the precise downstream pathways how mTOR regulates IL-12 responses by the p38 MAPK. J. Immunol. 185: 6205–6213. 17. Weichhart, T., and M. D. Sa¨emann. 2009. The multiple facets of mTOR in versus IL-10 needs further investigation. In this study, we fo- immunity. Trends Immunol. 30: 218–226. cused on delineating the mechanism of p38-depdendent mTOR 18. Thomson, A. W., H. R. Turnquist, and G. Raimondi. 2009. Immunoregulatory functions of mTOR inhibition. Nat. Rev. Immunol. 9: 324–337. regulation in myeloid immune cells. 19. Powell, J. D., K. N. Pollizzi, E. B. Heikamp, and M. R. Horton. 2012. Regulation Rapamycin is currently evaluated as adjuvant, especially of immune responses by mTOR. Annu. Rev. Immunol. 30: 39–68. because of its ability to enhance memory CD8+ T cell responses. In 20. Araki, K., A. H. Ellebedy, and R. Ahmed. 2011. TOR in the immune system. Curr. Opin. Cell Biol. 23: 707–715. addition, previous work also established that rapamycin exerts 21. Weintz, G., J. V. Olsen, K. Fru¨hauf, M. Niedzielska, I. Amit, J. Jantsch, J. Mages, immunostimulatory effects via the innate immune system that may C. Frech, L. Do¨lken, M. Mann, and R. Lang. 2010. The phosphoproteome of toll- contribute to the adjuvant properties of rapamycin (23–25, 48, 49). like receptor-activated macrophages. Mol. Syst. Biol. 6: 371. 22. Fukao, T., M. Tanabe, Y. Terauchi, T. Ota, S. Matsuda, T. Asano, T. Kadowaki, However, its immunosuppressive activity will likely prevent the T. Takeuchi, and S. Koyasu. 2002. PI3K-mediated negative feedback regulation inclusion of rapamycin in widely distributed . Our data of IL-12 production in DCs. Nat. Immunol. 3: 875–881. 23. Ohtani, M., S. Nagai, S. Kondo, S. Mizuno, K. Nakamura, M. Tanabe, suggest that inhibition of p38 might be similarly effective as ad- T. Takeuchi, S. Matsuda, and S. Koyasu. 2008. Mammalian target of rapa- juvant strategy where strong Th1 responses are desired, and more- mycin and glycogen synthase kinase 3 differentially regulate lipopolysaccha- over, it might avoid the potent immunosuppressive effects on the ride-induced interleukin-12 production in dendritic cells. 112: 635–643. 24. Weichhart, T., G. Costantino, M. Poglitsch, M. Rosner, M. Zeyda, K. M. Stuhlmeier, T cell compartment as p38 is regarded dispensable for T cell T. Kolbe, T. M. Stulnig, W. H. Ho¨rl, M. Hengstschla¨ger, et al. 2008. The TSC- function (50). Indeed, in a mouse model of Leishmania major mTOR signaling pathway regulates the innate inflammatory response. Immunity , vaccination with SB203580 was protective by inducing 29: 565–577. 25. Haidinger, M., M. Poglitsch, R. Geyeregger, S. Kasturi, M. Zeyda, G. J. Zlabinger, efficient Th1 immunity (16). B. Pulendran, W. H. Ho¨rl, M. D. Sa¨emann, and T. Weichhart. 2010. A versatile role Downloaded from In summary, we have identified and characterized a pathway of mammalian target of rapamycin in human dendritic cell function and differ- from p38a to mTOR via MK2 and TSC1/TSC2 in myeloid im- entiation. J. Immunol. 185: 3919–3931. 26. Weichhart, T., M. Haidinger, K. Katholnig, C. Kopecky, M. Poglitsch, mune cells that tunes the immune response according to envi- C. Lassnig, M. Rosner, G. J. Zlabinger, M. Hengstschla¨ger, M. Mu¨ller, et al. ronmental input signals. 2011. Inhibition of mTOR blocks the anti-inflammatory effects of glucocorti- coids in myeloid immune cells. Blood 117: 4273–4283. 27. Turnquist, H. R., J. Cardinal, C. Macedo, B. R. Rosborough, T. L. Sumpter, Acknowledgments D. A. Geller, D. Metes, and A. W. Thomson. 2010. mTOR and GSK-3 shape the We thank Margarethe Merio for excellent technical assistance. CD4+ T-cell stimulatory and differentiation capacity of myeloid DCs after ex- http://www.jimmunol.org/ posure to LPS. Blood 115: 4758–4769. 28. Jiang, Q., J. M. Weiss, T. Back, T. Chan, J. R. Ortaldo, S. Guichard, and Disclosures R. H. Wiltrout. 2011. mTOR kinase inhibitor AZD8055 enhances the immu- The authors have no financial conflicts of interest. notherapeutic activity of an agonist CD40 antibody in cancer treatment. Cancer Res. 71: 4074–4084. 29. Inoki, K., Y. Li, T. Zhu, J. Wu, and K. L. Guan. 2002. TSC2 is phosphorylated References and inhibited by Akt and suppresses mTOR signalling. Nat. Cell Biol. 4: 648– 657. 1. Kawai, T., and S. Akira. 2010. The role of pattern-recognition receptors in innate 30. Chen, W., T. Ma, X. N. Shen, X. F. Xia, G. D. Xu, X. L. Bai, and T. B. Liang. immunity: update on Toll-like receptors. Nat. Immunol. 11: 373–384. 2012. Macrophage-induced tumor angiogenesis is regulated by the TSC2-mTOR 2. Widmann, C., S. Gibson, M. B. Jarpe, and G. L. Johnson. 1999. Mitogen- pathway. Cancer Res. 72: 1363–1372. activated protein kinase: conservation of a three-kinase module from yeast to 31. Ohtani, M., T. Hoshii, H. Fujii, S. Koyasu, A. Hirao, and S. Matsuda. 2012. by guest on October 1, 2021 human. Physiol. Rev. 79: 143–180. Cutting edge: mTORC1 in intestinal CD11c+ CD11b+ dendritic cells regulates 3. Dong, C., R. J. Davis, and R. A. Flavell. 2002. MAP kinases in the immune intestinal homeostasis by promoting IL-10 production. J. Immunol. 188: 4736–4740. response. Annu. Rev. Immunol. 20: 55–72. 32. Pan, H., T. F. O’Brien, P. Zhang, and X. P. Zhong. 2012. The role of tuberous 4. Coulthard, L. R., D. E. White, D. L. Jones, M. F. McDermott, and S. A. Burchill. sclerosis complex 1 in regulating innate immunity. J. Immunol. 188: 3658–3666. 2009. p38(MAPK): stress responses from molecular mechanisms to therapeutics. 33. Zhang, H., G. Cicchetti, H. Onda, H. B. Koon, K. Asrican, N. Bajraszewski, Trends Mol. Med. 15: 369–379. F. Vazquez, C. L. Carpenter, and D. J. Kwiatkowski. 2003. Loss of Tsc1/Tsc2 5. Wagner, E. F., and A. R. Nebreda. 2009. Signal integration by JNK and p38 activates mTOR and disrupts PI3K-Akt signaling through downregulation of MAPK pathways in cancer development. Nat. Rev. Cancer 9: 537–549. PDGFR. J. Clin. Invest. 112: 1223–1233. 6. Cuadrado, A., and A. R. Nebreda. 2010. Mechanisms and functions of p38 34. Kwiatkowski, D. J., H. Zhang, J. L. Bandura, K. M. Heiberger, M. Glogauer, MAPK signalling. Biochem. J. 429: 403–417. N. el-Hashemite, and H. Onda. 2002. A mouse model of TSC1 reveals sex- 7. Lee, J. C., J. T. Laydon, P. C. McDonnell, T. F. Gallagher, S. Kumar, D. Green, dependent lethality from liver hemangiomas, and up-regulation of p70S6 ki- D. McNulty, M. J. Blumenthal, J. R. Heys, S. W. Landvatter, et al. 1994. A nase activity in Tsc1 null cells. Hum. Mol. Genet. 11: 525–534. protein kinase involved in the regulation of inflammatory cytokine biosynthesis. 35. Strobl, B., I. Bubic, U. Bruns, R. Steinborn, R. Lajko, T. Kolbe, M. Karaghiosoff, Nature 372: 739–746. U. Kalinke, S. Jonjic, and M. Mu¨ller. 2005. Novel functions of tyrosine kinase 2 8. Schett, G., J. Zwerina, and G. Firestein. 2008. The p38 mitogen-activated protein in the antiviral defense against murine cytomegalovirus. J. Immunol. 175: 4000– kinase (MAPK) pathway in rheumatoid arthritis. Ann. Rheum. Dis. 67: 909–916. 4008. 9. Soloaga, A., S. Thomson, G. R. Wiggin, N. Rampersaud, M. H. Dyson, 36. Ehlting, C., N. Ronkina, O. Bo¨hmer, U. Albrecht, K. A. Bode, K. S. Lang, C. A. Hazzalin, L. C. Mahadevan, and J. S. Arthur. 2003. MSK2 and MSK1 A. Kotlyarov, D. Radzioch, M. Gaestel, D. Ha¨ussinger, and J. G. Bode. 2011. mediate the mitogen- and stress-induced phosphorylation of histone H3 and Distinct functions of the mitogen-activated protein kinase-activated protein HMG-14. EMBO J. 22: 2788–2797. (MAPKAP) kinases MK2 and MK3: MK2 mediates lipopolysaccharide-induced 10.Lehner,M.D.,F.Schwoebel,A.Kotlyarov,M.Leist,M.Gaestel,and signal transducers and activators of transcription 3 (STAT3) activation by pre- T. Hartung. 2002. Mitogen-activated protein kinase-activated protein kinase 2- venting negative regulatory effects of MK3. J. Biol. Chem. 286: 24113–24124. deficient mice show increased susceptibility to Listeria monocytogenes infection. 37. Li, Y., K. Inoki, P. Vacratsis, and K. L. Guan. 2003. The p38 and MK2 kinase J. Immunol. 168: 4667–4673. cascade phosphorylates tuberin, the tuberous sclerosis 2 gene product, and en- 11. Hammaker, D., and G. S. Firestein. 2010. “Go upstream, young man”: lessons hances its interaction with 14-3-3. J. Biol. Chem. 278: 13663–13671. learned from the p38 saga. Ann. Rheum. Dis. 69(Suppl. 1): i77–i82. 38. Russell, R. C., C. Fang, and K. L. Guan. 2011. An emerging role for TOR 12. Pargellis, C., L. Tong, L. Churchill, P. F. Cirillo, T. Gilmore, A. G. Graham, signaling in mammalian tissue and stem cell physiology. Development 138: P. M. Grob, E. R. Hickey, N. Moss, S. Pav, and J. Regan. 2002. Inhibition of p38 3343–3356. MAP kinase by utilizing a novel allosteric binding site. Nat. Struct. Biol. 9: 268– 39. Clark, A., J. Dean, C. Tudor, and J. Saklatvala. 2009. Post-transcriptional gene 272. regulation by MAP kinases via AU-rich elements. Front. Biosci. 14: 847–871. 13. Kuma, Y., G. Sabio, J. Bain, N. Shpiro, R. Ma´rquez, and A. Cuenda. 2005. 40. Goh, K. C., M. J. deVeer, and B. R. Williams. 2000. The protein kinase PKR is BIRB796 inhibits all p38 MAPK isoforms in vitro and in vivo. J. Biol. Chem. required for p38 MAPK activation and the innate immune response to bacterial 280: 19472–19479. endotoxin. EMBO J. 19: 4292–4297. 14. Kim, C., Y. Sano, K. Todorova, B. A. Carlson, L. Arpa, A. Celada, T. Lawrence, 41. Thuraisingam, T., Y. Z. Xu, K. Eadie, M. Heravi, M. C. Guiot, R. Greemberg, K. Otsu, J. L. Brissette, J. S. Arthur, and J. M. Park. 2008. The kinase p38 alpha M. Gaestel, and D. Radzioch. 2010. MAPKAPK-2 signaling is critical for cu- serves cell type-specific inflammatory functions in skin injury and coordinates taneous . J. Invest. Dermatol. 130: 278–286. pro- and anti-inflammatory gene expression. Nat. Immunol. 9: 1019–1027. 42. Perdiguero, E., P. Sousa-Victor, V. Ruiz-Bonilla, M. Jardı´, C. Caelles, 15. Guo, X., R. E. Gerl, and J. W. Schrader. 2003. Defining the involvement of A. L. Serrano, and P. Mun˜oz-Ca´noves. 2011. p38/MKP-1-regulated AKT co- p38alpha MAPK in the production of anti- and proinflammatory cytokines using ordinates macrophage transitions and resolution of inflammation during tissue an SB 203580-resistant form of the kinase. J. Biol. Chem. 278: 22237–22242. repair. J. Cell Biol. 195: 307–322. The Journal of Immunology 9

43. Matzinger, P., and T. Kamala. 2011. Tissue-based class control: the other side of 47. Rane, M. J., P. Y. Coxon, D. W. Powell, R. Webster, J. B. Klein, W. Pierce, tolerance. Nat. Rev. Immunol. 11: 221–230. P. Ping, and K. R. McLeish. 2001. p38 Kinase-dependent MAPKAPK-2 acti- 44. Cully, M., A. Genevet, P. Warne, C. Treins, T. Liu, J. Bastien, B. Baum, N. Tapon, vation functions as 3-phosphoinositide-dependent kinase-2 for Akt in human S. J. Leevers, and J. Downward. 2010. A role for p38 stress-activated protein kinase neutrophils. J. Biol. Chem. 276: 3517–3523. in regulation of cell growth via TORC1. Mol. Cell. Biol. 30: 481–495. 48. Janes, M. R., and D. A. Fruman. 2009. Immune regulation by rapamycin: 45. Wu, X. N., X. K. Wang, S. Q. Wu, J. Lu, M. Zheng, Y. H. Wang, H. Zhou, moving beyond T cells. Sci. Signal. 2: pe25. H. Zhang, and J. Han. 2011. Phosphorylation of Raptor by p38beta participates 49. Jagannath, C., D. R. Lindsey, S. Dhandayuthapani, Y. Xu, R. L. Hunter, Jr., in arsenite-induced mammalian target of rapamycin complex 1 (mTORC1) ac- and N. T. Eissa. 2009. Autophagy enhances the efficacy of BCG vaccine by tivation. J. Biol. Chem. 286: 31501–31511. increasing peptide presentation in mouse dendritic cells. Nat. Med. 15: 267– 46. Zheng, M., Y. H. Wang, X. N. Wu, S. Q. Wu, B. J. Lu, M. Q. Dong, H. Zhang, 276. P. Sun, S. C. Lin, K. L. Guan, and J. Han. 2011. Inactivation of Rheb by PRAK- 50. Kim, J. M., J. M. White, A. S. Shaw, and B. P. Sleckman. 2005. MAPK p38 mediated phosphorylation is essential for energy-depletion-induced suppression alpha is dispensable for lymphocyte development and proliferation. J. Immunol. of mTORC1. Nat. Cell Biol. 13: 263–272. 174: 1239–1244. Downloaded from http://www.jimmunol.org/ by guest on October 1, 2021