Immunobiology 219 (2014) 315–322

Contents lists available at ScienceDirect

Immunobiology journal homepage: www.elsevier.com/locate/imbio

Dimethyl sulfoxide inhibits NLRP3 inflammasome activation Huijeong Ahn a , Jeeyoung Kim a , Eui-Bae Jeung b , Geun-Shik Lee a,∗ a

College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon 200-701, Republic of Korea Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju 361-763, Republic of Korea b

a r t i c l e

i n f o

Article history: Received 30 July 2013 Received in revised form 16 October 2013 Accepted 14 November 2013 Available online 22 November 2013 Keywords: Cytokine DMSO Inflammasome Interleukin-1 Macrophages NLRP3

a b s t r a c t Dimethyl sulfoxide (DMSO) is an amphipathic molecule that is commonly/widely used as a solvent for biological compounds. In addition, DMSO has been studied as a medication for the treatment of inflammation, cystitis, and arthritis. Based on the anti-inflammatory characteristics of DMSO, we elucidated the effects of DMSO on activation of inflammasomes, which are cytoplasmic multi-protein complexes that mediate the maturation of interleukin (IL)-1␤ by activating caspase-1 (Casp1). In the present study, we prove that DMSO attenuated IL-1␤ maturation, Casp1 activity, and ASC pyroptosome formation via NLRP3 inflammasome activators. Further, NLRC4 and AIM2 inflammasome activity were not affected, suggesting that DMSO is a selective inhibitor of the NLRP3 inflammasomes. The anti-inflammatory effect of DMSO was further confirmed in animal, LPS-endotoxin sepsis and inflammatory bowel disease models. In addition, DMSO inhibited LPS-mediating IL-1s transcription. Taken together, DMSO shows anti-inflammatory characteristics, attenuates NLRP3 inflammasome activation, and mediates inhibition of IL-1s transcription. Crown Copyright © 2013 Published by Elsevier GmbH. All rights reserved.

Introduction Dimethyl sulfoxide (DMSO, (CH3 )2 SO) is an amphipathic molecule with a highly polar domain and two apolar groups, making it soluble in both aqueous and organic media (Santos et al., 2003). Due to its physical and chemical properties, DMSO is a very efficient solvent for water-insoluble compounds and is a hydrogen-bound disrupter. Although the biological effects of DMSO have not been fully identified, it is commonly adopted as a solvent of pharmacological studies. DMSO was previously reported to increase drug permeability and transport across tissue membranes using animal models (Jacob and Herschler, 1986). Thus, DMSO is popularly used as a vehicle for drug therapy for treatment of various diseases, including dermatological disorders, amyloidosis, gastrointestinal diseases, traumatic brain edema, musculoskeletal disorders, pulmonary adenocarcinoma, rheumatologic diseases, and schizophrenia (Santos et al., 2003). DMSO has been reported as a hydroxyl radical scavenger that improves mitochondrial oxidative phosphorylation and neutralizes the cytotoxic effects of free radicals formed upon ischemia in mitochondria (Colucci et al., 2008). DMSO has also been studied as a treatment for inflammation, cystitis, and arthritis (Kloesch et al., 2011).

∗ Corresponding author at: College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chucheon 200-701, Republic of Korea. Tel.: +82 33 250 8683; fax: +82 33 244 2367. E-mail address: [email protected] (G.-S. Lee).

DMSO exhibits anti-inflammatory properties by reducing NF-␬B activation in association with decreased secretion and/or mRNA expression of pro-inflammatory mediators such as interleukin (IL)1␤ (Hollebeeck et al., 2011). However, there are conflicting reports about the effect of DMSO on IL-1␤ production (Xing and Remick, 2005). Inflammation mediated by the innate immune system as the first line of host defense against pathogenic microbial or pathogen infection is an acute response that limits tissue damage and harm in the body (Schroder et al., 2010). Membrane-associated Toll-like receptors (TLR), c-type lectin receptors (CLR), cytosolic nucleotide-binding domain and leucine-rich repeat receptors (NLRs), retinoic acid-inducible gene (RIG)-like helicase (RLH) receptors, and absent in melanoma (AIM)-like receptors comprise the five main pattern recognition receptor families. TLR activation triggers different signaling cascades that result in NF-␬B activation and the synthesis of various pro-inflammatory mediators and cytokines (Medzhitov, 2001). One pro-inflammatory cytokine, IL1␤, is an important inflammatory mediator generated at sites of injury or immunological challenge that coordinates programs as diverse as cellular recruitment to sites of infection or injury as well as the regulation of sleep, appetite, and body temperature (Dinarello et al., 2010). However, dysregulation and chronic inflammation may result in secondary damage and immune pathology to the host. IL-1␤ activity is rigorously controlled at the levels of expression, maturation, and secretion. Pro-inflammatory stimuli induce expression of the inactive pro-from of IL-1␤, but its maturation and release are controlled by inflammasomes.

0171-2985/$ – see front matter. Crown Copyright © 2013 Published by Elsevier GmbH. All rights reserved. http://dx.doi.org/10.1016/j.imbio.2013.11.003

316

H. Ahn et al. / Immunobiology 219 (2014) 315–322

Inflammasomes are multiprotein complexes that operate as platforms for the activation of caspase-1 and are composed of one of several NLR and PYHIN proteins, including NLRP1, NLRP3, NLRC4, and AIM2 (Rathinam et al., 2012). Inflammasomes are sensors of endogenous or exogenous pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs), which govern cleavage of effector pro-inflammatory cytokines such as pro-IL-1␤ and pro-IL-18. Inflammasomes have been found to regulate other important aspects of inflammation and tissue repair such as pyroptosis, a form of cell death (Lamkanfi and Dixit, 2011; Lamkanfi, 2011). Mutation of just one inflammasomeassociating gene has been shown to induce auto-inflammatory disease (McDermott et al., 1999). Inflammasome activation requires two steps, priming and activation. During the priming step, TLR stimuli induce production of pro-IL-1␤ and inflammasome components such as NLRP3 (Lee et al., 2012). In the activation step, inflammasome activators facilitate assembly of inflammasome complexes to induce self-cleavage of caspase-1. Although the effect of DMSO on IL-1␤ production has been progressively studied, controversial results have been reported (Xing and Remick, 2005). In this study, we investigated the effects of DMSO on the activation of well-described inflammasomes such as NLRP3, NLRC4, and AIM2 using bone marrow-derived macrophages (BMDMs) and animal models.

(Sigma–Aldrich Co.) were collected for further analysis. For cytotoxicity assay, BMDMs (10,000 cells/well) were plated in a 96-well plate (SPL life science Co.) and allowed to be attached for 3 h. The BMDMs were treated with the indicated dosage of DMSO for 1 h and replaced with fresh media (RPMI 1640 containing 10% FBS and antibiotics) followed by further incubation for 14 h. The cytotoxicity was measured by Cell Counting Kit-8 (Dojindo Molecular Technologies, Inc. MD, USA) as manufacture’s protocol.

Materials and methods

Bacteria growth

Differentiation of bone marrow-derived macrophages (BMDMs)

S. typhimurium (Sun and Hahn, 2012) were kindly provided from Prof. Tae-Wook Han (Kangwon National University, Gangwon-do, Republic of Korea). Salmonella were grown on Luria-Bertani (LB, Laboratories Conda, Madrid, Spain) agar plates for 18 h at 37 ◦ C. A single colony was transferred to LB broth, after which the culture was incubated for 18 h with shaking at 37 ◦ C. The cultured bacteria were serially diluted and plated on the agar plates to calculate colony-forming units (cfu). To elucidate the effect of DMSO on bacterial growth, Salmonella (1.5 × 104 cfu) were growth in LB broth containing PBS, DMSO, or gentamycin for 18 h at 37 ◦ C, followed by calculation of cfu. For Salmonella host invasion assay, BMDMs (1 × 106 cells per well) were plated in 12-well plates and inoculated with Salmonella (2 × 107 cfu) in DMEM containing 10% FBS for 1 h. The plate was then washed with Gentamycin (50 ␮g/ml; Komipharm International Co., Ltd., Gyeonggi-do, Republic of Korea) containing PBS to eliminate extracellular Salmonella and then plated on an LB plate to calculate cfu.

Unless otherwise indicated, all materials for cell culture were obtained from PAA (GE Healthcare Bio-Sciences Co., NJ, USA). BMDMs were obtained by differentiating bone marrow progenitors from the tibia and femur bones using L929 cell-conditioned medium (LCCM) as a source of granulocyte/macrophage colonystimulating factor (Englen et al., 1995). The progenitors were cultured in RPMI 1640 supplemented with 10% fetal bovine serum (FBS), 30% LCCM, 100 U/ml of penicillin, and 100 ␮g/ml of streptomycin. Cells were seeded in non-tissue culture-treated Petri dishes (SPL Life Science Co., Gyeonggi-do, Republic of Korea) and incubated at 37 ◦ C in a 5% CO2 atmosphere for 7 days. Cell treatments BMDMs (1.0 × 106 cells per well) were plated in 12-well plates (SPL Life Science Co.) and primed with 10 ␮g/ml of lipopolysaccharide (LPS; Sigma–Aldrich Co., MO, USA) in RPMI 1640 containing 10% FBS and antibiotics for 3 h. After LPS priming, BMDMs were subjected to the following activation step for 1 or 3 h. For NLRP3 inflammasome activation, the medium was replaced with RPMI 1640 supplemented with ATP (3.5 mM; InvivoGen, CA, USA) for 1 h, nigericin (NG, 40 ␮M; Tocris Bioscience, Bristol, UK) for 1 h, calcium pyrophosphate dihydrate (CPPD; 200 ␮g/ml; InvivoGen) for 1 h, CaCl2 (1 mM; Biosesang, Seoul, Republic of Korea) for 1 h, aluminum potassium sulfate (Alum; 200 ␮g/ml; Daejeung Chemicals & Materials Co., Gyeonggi-do, Republic of Korea) for 3 h. For NLRC4 inflammasome activation, the media were exchanged with Salmonella typhimurium (OD600: 1.2, 0.5%) or purified flagellin (500 ng/ml; InvivoGen) with Lipofectamin 10 ml/ml (Invitrogen, CA, USA) and collected after 1 h. For AIM2 inflammasome activation, LPS-primed BMDMs were supplemented with 2 ␮g/ml of dsDNA containing 4 ␮l/ml of Lipofectamine 2000 (Invitrogen) for 1 h. To determine the inhibitory effect of DMSO on inflammasome activation, DMSO (Daejeung Chemicals & Materials Co.) was co-treated with the above activators. Cellular supernatant (Sup), lysate (Lys), and cross-linked pellets (Pellet) with suberic acid bis

Western blot analysis Sup, Lys, and Pellet samples were separated by SDS-PAGE (10% or 16%) and blotted onto a polyvinylidene difluoride membrane (Pall Co., NY, USA). Immunoblots were probed overnight at 4 ◦ C with anti-mouse IL-1␤ antibody (AF-401-NA, R&D Systems, MN, USA), anti-caspase-1 p20 antibody (06-503, EMD Millipore Co, MA, USA), anti-ASC antibody (sc-22514, Santa Cruz Biotechnology, CA, USA), anti-caspase-1 antibody (sc-622, Santa Cruz Biotechnology), or anti-actin antibody (sc-1615, Santa Cruz Biotechnology). The membranes were further probed with HRP-conjugated 2nd antisera (sc-2020 or sc-2004, Santa Cruz Biotechnology) and visualized by Power-Opti ECLTM solution (BioNote Co., Gyeonggi-do, Republic of Korea) and a cooled CCD camera System (AE-9150, EZ-Capture II, ATTO technology, Tokyo, Japan).

Animal experiments C57BL/6 mice (6- to 8-weeks-old) were purchased from Orient Co. (Seongnam, Republic of Korea) or Narabio Co. (Seoul, Republic of Korea). The animals were housed at 24 ± 1 ◦ C under 50% humidity and a 12-h light/dark cycle. All animal experiments were carried out in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals and approved by the Institutional Animal Care and Use Committee of Kangwon National University (ACUCC; approval no. KIACUC-12-0009). For LPS-induced endotoxin shock, mice were intraperitoneally (ip) pre-treated with vehicle (200 ␮l of saline) or 10% DMSO at 1 h and 1 day before LPS (20 mg/kg) ip injection. The animals were observed every 8 h for 3 days. To induce inflammatory bowel disease (IBD) by dextran sulfate sodium (DSS), mice drank tap water only or water containing 3% DSS (MWs = 40 kDa; MP Biomedicals, CA, USA) for 7 days. The mice were then ip injected with 10% DMSO in 200 ␮l of saline on days 0, 2, 4, 6, and 8. Body weights were recorded daily. For the Salmonella-mediated lethality study, mice were ip injected with vehicle (200 ␮l of saline) or 10% DMSO at

H. Ahn et al. / Immunobiology 219 (2014) 315–322

1 h and 1 day before oral S. typhimurium (3 × 106 cfu per mouse) administration. Lethality was monitored every 8 h for 9 days.

317

NM 007393) 5 -AGC CAT GTA CGT AGC CAT CC-3 and 5 -CTC TCA GCT GTG GTG GTG AA-3 . Statistical analyses

RNA extraction and real-time PCR For RNA extraction, BMDMs (2.0 × 106 cells per well) were plated in 6-well plates (SPL Life Science Co.) and treated 10 ng/ml of LPS with the indicated DMSO for 3 h. Total RNA was extracted using Trizol (Invitrogen) and reverse-transcribed to first-stand complementary DNA (cDNA) using random primer (9-mer) and m M-MLV reverse transcripatase (Invitrogen) according to the manufacturer’s protocols. Transcripts were quantitated using SyberGreen (Applied Biosystems, Carlsbad, CA, USA) and the Eco Real-Time PCR system (Illumina, San Diego, CA, USA). Quantitation was normalized with beta-actin (Actb). IL-1␤ (Il1b; Genebank ID: NM 008361) primers 5 -CCC AAG CAA TAC CCA AAG AA-3 and 5 -GCT TGT GCT CTG CTT GTG AG-3 ; IL-1␣ (Il1a; NM 010554) 5 -CCG ACC TCA TTT TCT TCT GG-3 and 5 -GTG CAC CCG ACT TTG TTC TT-3 ; IL-6 (Il6; NM 031168) 5 -CCG GAG AGG AGA CTT CAC AG-3 and 5 -TCC ACG ATT TCC CAG AGA AC-3 ; TNF-␣ (Tnfa; NM 013693) 5 -ACG GCA TGG ATC TCA AAG AC-3 and 5 -GTG GGT GAG GAG CAC GTA GT-3 ; IL-10 (Il10; NM 010548) 5 -GCC TGG CTC AGC ACT GCT AT-3 and 5 GAA GGC AGT CCG CAG CTC TA-3 ; IL-12␣ (Il12a; NM 001159424) 5 -GCT ATC TGA GCT CCG CCT GA-3 and 5 -AGG AGC TTA AGG CCC ACC AG-3 ; IL-1RN (Il1rn; NM 001039701) 5 -GCC TGA TCA CTC TGG CCA TC-3 and 5 -AGG CCA GCC AAC AGA CTG AG-3 ; ␤-actin (Actb;

Statistical analyses were performed with a t-test for the two groups, or one-way ANOVA for multiple groups, and survival analysis for lethality test using GraphPad Prism (GraphPad Software, San Diego, CA). Results DMSO inhibits inflammasome activation To elucidate the effects of DMSO on inflammasome activation, we firstly treated various dosages of DMSO to LPS-primed BMDMs with/without ATP, a specific activator of the NLRP3 inflammasome. As shown in Fig. 1A, DMSO alone did not induce IL-1␤ (p17, active form) production nor alter pro-IL-1␤ secretion in the cell supernatant (Sup). In addition, any cytotoxicity of BMDMs was not detected by DMSO treatment (up to 10%) based on the cytotoxicity assay (Fig. 1B), and unchanged proIL-1␤ and actin expressions. However, DMSO co-treatment with ATP promoted reduction of IL-1␤ secretion in a dose-dependent manner. Active IL-1␤ was no longer present in the lysate (Lys), indicating that DMSO inhibited IL-1␤ production but not secretion. We further treated the other NLRP3 inflammasome

Fig. 1. Effect of DMSO on inflammasome activation. BMDMs (1 × 106 cells per well) were primed by LPS (10 ␮g/ml) in RPMI medium containing 10% FBS and antibiotics for 3 h. (A) Cells were supplied with RPMI medium in the present with DMSO with/without ATP (3.5 mM) and were analyzed for IL-1 ␤ secretion by immunoblotting. (B) The cytotoxicity of DMSO was measured after applying the indicated dosage of DMSO on BMDMs for 1 h like to the inflammasome activating step. Triton X-100 (0.01%, Triton) suggested by the manufacturer presents complete cell death. Triton treated group sets as 0% of the survival rate and non-treated group sets as 100%. Data represent the mean ± s.e.m of three independent experiments, each performed in triplicate. (C) LPS-primed BMDMs were treated with NG (40 ␮M), CPPD (200 ␮g/ml), and CaCl2 (1 mM) for 1 h, or Alum (200 ␮g/ml) for 3 h in the present of DMSO. (D) LPS-primed BMDMs were activated in the present of DMOS with Salmonella (OD600: 1.2, 0.5%) or 2 ␮g/ml of dsDNA with 4 ␮l/ml of Lipofectamine 2000 for 1 h. Cell culture supernatants (Sup) and cell lysates (Lys) were analyzed by immunoblotting as indicated.

318

H. Ahn et al. / Immunobiology 219 (2014) 315–322

Fig. 2. DMSO inhibits NLRP3 inflammasome activation. (A) LPS-primed BMDMs were co-treated with NLRP3 inflammasome activators, ATP or nigericin (NG), as well as the indicated percentages of DMSO. Cell culture supernatants (Sup), cell lysates (Lys), and cross-linked pellets (Pellet) from whole-cell lysates were analyzed by immunoblotting as indicated. (B) LPS-primed BMDMs (1.5 × 106 cells per reaction) were further incubated with NG for 5 min and then subjected to Casp-1 activity assay using YVAD-pNA. Data represent the mean ± s.e.m for three independent experiments. (C) Mice (n = 10 per each group) were intraperitoneally (ip) injected with saline (200 ␮l) with or without 10% DMSO at 1 h before LPS (20 mg/kg) administration. Survival rates were observed at the indicated times. (D) Mice (n = 10 per each group) were treated with 3% DSS for the indicated period (DSS) and then ip injected with 10% DMSO in saline (200 ␮l) at the indicated time (arrows). Body weights were monitored daily for 11 days. Data represent the mean ± s.d. for five mice per group. *P < 0.05 vs. DSS only.

activators, such as nigericin (NG), calcium pyrophosphate dihydrate (CPPD), calcium chloride (CaCl2 ), and aluminum potassium sulfate (Alum), to LPS-primed BMDMs in the presence of DMSO to confirm the inhibitory effect of DMSO on NLRP3 inflammasome activation. DMSO attenuated mature IL-1␤ production in NG, CPPD, CaCl2 , and Alum-treated macrophages, whereas the inhibitory potency of DMSO on NLRP3 inflammasome activation varied (Fig. 1C). To confirm the inhibitory potencies of DMSO on the NLRC4 and AIM2 inflammasomes, we co-treated DMSO to Salmonella-infected and dsDNA-transfected BMDMs, respectively (Fig. 1D). Salmonella-mediated IL-1␤ maturation via the NLRC4 inflammasome was attenuated by DMSO, similar to the NLRP3 inflammasome result. However, dsDNA-induced IL-1␤ maturation via the AIM2 inflammasome was not altered by DMSO, suggesting that DMSO only inhibited the NLRP3 and NLRC4 inflammasomes. Hence, we conclude that DMSO could be a candidate for the selective inhibition of the NLRP3 and NLRC4 inflammasomes.

DMSO inhibits NLRP3 inflammasome Inflammasome activation involves the production of activated caspase-1 (Casp1; p10 and p20) as well as the induction of pyroptosis (Lamkanfi and Dixit, 2012). To determine the role of DMSO in these two phenomena of inflammasome activation, we observed the effects of DMSO on secretion of Casp1 (p20) in the cell supernatant as well as on formation of the ASC pyroptosome in the insoluble pellet after cross-linking with disuccinimidyl suberate (Fig. 2A). Secretion of Casp1 was attenuated by DMSO in the presence of ATP or NG, although pro-Casp1 was evenly observed in the lysate. In addition, DMSO reduced formation of the ASC pyroptosome by NLRP3 inflammasome activators. Thus, DMSO inhibited Casp1 secretion and ASC pyroptosis followed by NLRP3 inflammasome activation, similar to the results on IL-1␤ secretion. In addition, we tested Casp1 activity by incubating YVAD-pNA, a Casp1 substrate, in BMDM lysate containing NG with or without DMSO (Fig. 2B). Casp1

H. Ahn et al. / Immunobiology 219 (2014) 315–322

activity was induced by NG alone, whereas activities were inhibited by DMSO in a dose-dependent manner. Taken together, we propose that DMSO blocked activation of the NLRP3 inflammasome. In the literature, the susceptibility of LPS-induced endotoxin shock is reduced in NLRP3 gene-depleted mice when compared with intact mice (Mariathasan et al., 2006). In addition, loss of body weight in an inflammatory bowel disease (IBD) model was shown to be less severe in NLRP3-deficient mice (Bauer et al., 2010). In this study, we elucidated the protective effects of DMSO in these animal models. As shown in Fig. 2C, mice treated with LPS alone presented a 30% of survival rate at 32 h after post-injection, whereas 70% of mice were alive in the DMSO-treated group at the same time point. Finally, DMSO-treated mice showed 60% survival by the end of the experiment. In the IBD model, mice that received water containing dextran sulfate sodium (DSS) presented 13.5% body weight loss, whereas DMSO-injected mice treated with DSS-containing water (DSS + DMSO) showed only 6.7% body weight loss at the same time point (Fig. 2D). Those mice treated with tap water or DMSO only showed no body weight loss. Thus, DMSO ameliorated the severity of NLRP3 inflammasome-mediated disease symptoms in animals, which is similar to our result that DMSO inhibited IL-1␤ maturation in BMDMs. DMSO does not directly inhibit NLRC4 inflammasome DMSO not only inhibited IL-1␤ maturation via the NLRP3 inflammasome but also attenuated IL-1␤ production via the NLRC4 inflammasome, as seen in Fig. 1C. For more precise analysis of inflammasome activation, we elucidated formation of the ASC pyroptosome in the presence of DMSO with Salmonella for NLRC4 inflammasome activation or dsDNA for AIM2 inflammasome activation (Fig. 3A). As expected, DMSO attenuated formation of the ASC pyroptosome in the presence of Salmonella, similar to the results on IL-1␤ secretion. However, DMSO did not reduce formation of the ASC pyroptosome via AIM2 inflammasome activation. We further elucidated the inhibitory effect of DMSO on NLRC4 inflammasome activation in animals. Mice were treated with DMSO before oral administration of Salmonella, and the survival rate was observed (Fig. 3B). Although the infected mouse group treated with DMSO presented a bit higher survival rate than the group without DMSO treatment, there was no significant different between them. This result demonstrates that DMSO had no protective effect in mice against oral Salmonella infection. To elucidate the difference between in vivo and in vitro data, we further tested whether or not DMSO has a bactericidal effect on S. typhimurium, an activator of the NLRC4 inflammasome in the current study (Fig. 3C). DMSO did not show any bactericidal effect, whereas the antibiotic gentamycin completely blocked growth of Salmonella. Salmonella need to invade the cytoplasm for activation of inflammasomes due to their identity as cytosolic pattern recognition receptors. Thus, we elucidated the invasion of Salmonella into BMDMs as well as its intracellular growth rate in the presence of DMSO. After 1 h of inoculation of Salmonella to BMDMs, the number of intracellular bacteria was significantly reduced by DMSO, which implies that DMSO may have attenuated the penetrating property of Salmonella (Fig. 3D). In addition, we elucidated the effect of DMSO on NLRC4 inflammasome activation using the transfection of purified flagellin into BMDMs (Fig. 3E). Flagellin-mediated IL-1␤ secretion did not changed by DMSO. This result demonstrates that DMSO inhibited Salmonella-mediated IL-1␤ secretion by attenuating Salmonella invasion. DMSO reduces transcriptional levels of IL-1s and its antagonist DMSO has been studied as an anti-inflammatory medication due to its reported role as a free radical scavenger (Colucci et al.,

319

2008). Indeed, several studies have suggested that DMSO reduces the expression of pro-inflammatory cytokines via inhibition of NF-␬B signaling (Xing and Remick, 2005). However, there are conflicting reports about its effect on cytokine expression (Xing and Remick, 2005). Hence, we tested the effects of DMSO on the transcriptional levels of cytokines in BMDMs with or without LPS stimulation. DMSO dose-dependently reduced the expression of pro-inflammatory cytokines (IL-1␣, pro-IL-1␤, and IL-6) in BMDMs stimulated by LPS, although DMSO alone did not induce any pro-inflammatory cytokines (Fig. 4A–D). DMSO also attenuated the mRNA expression of IL-1RN (IL-1 receptor antagonist) (Fig. 4C). However, DMSO did not alter transcriptional levels of other cytokines, including TNF-␣, IL-12A, and IL-10 (Fig. 4F–H). Collectively, DMSO inhibited the transcription of IL-1s, IL-6, and IL-RN. Discussion Using in vitro and in vivo approaches, we provide evidence that DMSO plays an inhibitory role in NLRP3 inflammasome activation. Indeed, the anti-inflammatory properties of DMSO have been progressively studied (Loppnow and Libby, 1990; Reimann et al., 1994; Xaus et al., 2000), although there has been no investigation into inflammasome activation producing active IL-1␤. In this study, we elucidated the effect of DMSO on IL-1␤ production in the context of two distinct signals: the first signal leads to transcriptional upregulation and synthesis of pro-IL-1␤ while the second signal leads to IL-1␤ maturation and secretion through inflammasome activation. In the first signal step, DMSO down-regulated pro-IL-1␤ transcription, which may have attenuated secretion of IL-1␤ due to an insufficient amount of substrate for inflammasome activation. In the second signal step, DMSO inhibited IL-1␤ maturation and secretion even though cells were treated with the first signal. Thus, DMSO has dual properties that inhibit IL-1␤ production. In addition, the anti-inflammatory role of DMSO was confirmed using animal models. Although we have not fully defined the inhibitory mechanism of DMSO on inflammasome activation in the current study, we can speculate based on previous reports. DMSO is a well-known antioxidant that acts as a free radical scavenger (Santos et al., 2003; Phillis et al., 1998). Various models of NLRP3 inflammasome activation have been proposed, and the concept of reactive oxygen species (ROS) being upstream of NLRP3 activation has gained particular attention (Schroder et al., 2010). Genetic and pharmacological inhibition of NADPH oxidase-dependent ROS production is suggested to be upstream of NLRP3 inflammasome activation (Dostert et al., 2008). In addition, inhibitors of ROS production or ROS scavengers have been shown to strongly inhibit NLRP3 inflammasome activation (Cassel et al., 2008). Indeed, in line with the notion that mitochondria constitute the biggest source of cellular ROS, it was subsequently shown that mitochondria are the site of ROS production during NLRP3 inflammasome activation (Nakahira et al., 2011; Zhou et al., 2011). In this same context, it has also been demonstrated that inhibitors of mitochondrial ROS production (Nakahira et al., 2011) as well as knockdown of mitochondrial respiration mediated by reduced expression of voltage-dependent anion channels (Zhou et al., 2011) downregulate NLRP3-mediated inflammasome activation. Based on these findings, we suggest that DMSO is an ROS scavenger resulting in inhibition of NLRP3 inflammasome activation. ROS also activates pro-inflammatory gene expression in various innate immune signaling pathways (van de van de Veerdonk et al., 2010; Bulua et al., 2011). Bacterial LPS is commonly adopted as the first signal to induce pro-IL-1␤. LPS regulates several macrophage functions (Hsu and Twu, 2000; van Lenten and Fogelman, 1992), including the induction of inflammatory cytokines such as IL-1,

320

H. Ahn et al. / Immunobiology 219 (2014) 315–322

Fig. 3. Effect of DMSO on Salmonella mediating NLRC4 inflammasome activation. (A) LPS-primed BMDMs were co-treated with NLRC4 or AIM2 inflammasome activators, Salmonella, or dsDNA with Lipofectamine 2000 as well as the indicated percentages of DMSO. Cell culture supernatants (Sup), cell lysates (Lys), and cross-linked pellets (Pellet) from whole-cell lysates were analyzed by immunoblotting as indicated. (B) 8-weekold female mice (C57BL/6, n = 10 per each group) were intraperitoneally (ip) injected with saline (200 ␮l) with or without 10% DMSO at 1 h before Salmonella administration. Survival rates were observed at the indicated times. (C) Bactericidal effect of DMSO was measured in the presence of the indicated percentages of DMSO. Data represent the mean ± s.e.m for three independent experiments. (D) Invasion of Salmonella into BMDMs was analyzed in the presence of the indicated percentages of DMSO. Data represent cfu for six independent experiments. (E) LPS-primed BMDMs were transfected the purified flagellin in the present of DMSO and IL-1␤ secretion were analyzed.

IL-6, and TNF (Loppnow and Libby, 1990; Reimann et al., 1994; Xaus et al., 2000; Zhu et al., 2000). After LPS interacts with the TLR4 complex, macrophages produce reactive oxygen intermediates (Mendez et al., 1995) while nuclear factor (NF)-␬B activation is triggered, resulting in IL-1␤ production by macrophages (Xing and Remick, 2005). Further, DMSO has an inhibitory effect on NF-␬B activation (Chang et al., 1999). Hence, DMSO inhibits proinflammatory cytokine expression via ROS scavenging and NF-␬B inhibition, as seen in Fig. 4. Recently, we and others have reported that an increase in intracellular calcium triggers NLRP3 inflammasome activation (Lee et al., 2012; Murakami et al., 2012; Rossol et al., 2012). That is, inflammasome activators in the second signal step induce intracellular calcium ions, thereby facilitating formation of inflammasome complexes. DMSO has been reported to reduce the intracellular calcium concentration following exposure to hypoxic conditions and ionophore-induced calcium loading in several cell types (Santos et al., 2003). Although the precise mechanism by which DMSO prevents intracellular calcium induction has not been elucidated, it could explain how DMSO inhibits NLRP3 inflammasome activation. The explanation for the inhibitory effect of DMSO on NLRC4 inflammasome activation could involve inhibition of Salmonella host cell invasion by DMSO (Antunes et al., 2010). As shown in Fig. 3C, the invasion of Salmonella into BMDMs was attenuated by DMSO, although we could not explain the molecular aspects. The invasion of host cells by Salmonella is controlled by a complex cascade of regulatory proteins. One of the major players in the regulation of invasion-related gene expression is the OmpR/ToxRtype transcriptional regulator HilA (Lee et al., 1992). DMSO is reduced to dimethyl sulfide (DMS) during anaerobic growth, and

DMS strongly inhibits HilA gene expression, which may support our results (Antunes et al., 2010). We conclude that DMSO is an inhibitor of NLRP3 inflammasome activation in the current study. However, DMSO is a commonly adopted solvent and cryoprotectant in biological research. Indeed, several inflammasome activators such as nigericin, m-M3FBS, R528, Rotenone, and thapsigargin, have been dissolved in DMSO to activate inflammasomes (Lee et al., 2012; Zhou et al., 2011). In addition, DMSO is also used as a solvent for inhibitors of inflammasome activation such as BAPTA-AM (Murakami et al., 2012). Nevertheless, DMSO as a solvent does not interrupt inflammasome activation or inhibition as its final concentration is less than 1%. Thus, we suggest that a greater than 2% concentration of DMSO would have inhibitory effects on inflammasome activation and may present anti-inflammatory properties. Indeed, less than 10% of DMSO is well-adopted for cryopreservation and do not induce any cytotoxicity (Da Violante et al., 2002). Although the biological and pharmacological effects of DMSO have not been clearly defined, it is used extensively in a variety of fields. Due to its anti-inflammatory and ROS scavenger activities, DMSO is popularly used as a vehicle in drug therapy for various diseases, including dermatological disorders, amyloidosis, gastrointestinal diseases, traumatic brain edema, mausculoskeletal disorders, pulmonary adenocarcinoma, rheumatologic disease, schizophrenia, and Alzhimer’s diseases (Santos et al., 2003). On the other hand, several undesirable side effects stemming from the use of DMSO have been reported, namely cardiac problems and a garlic-like breath odor and taste in mice (Santos et al., 2003). In this study, we suggest an anti-inflammatory property for DMSO

H. Ahn et al. / Immunobiology 219 (2014) 315–322

321

Fig. 4. DMSO inhibits pro-inflammatory cytokine expressions. BMDMs (2 × 106 cells per well for RNA [A, C, D, E, F, G, and H] and 1 × 106 cells per well for protein [B]) were treated with LPS (10 ng/ml; w/LPS) supplemented with the indicated percentages of DMSO in RPMI medium containing 10% FBS and antibiotics for 3 h. Total RNA (1 ␮g) was isolated and cDNA synthesized using MMLV reverse transcriptase as described in Material and Methods. Relative levels of the indicated transcripts were analyzed using SYBR green-based quantitative real-time PCR. Data represent the mean ± s.e.m for three independent experiments. *P < 0.01 vs. LPS only.

322

H. Ahn et al. / Immunobiology 219 (2014) 315–322

via its inhibition of NLRP3 inflammasome activation as well as proinflammatory cytokine expression both in vitro and in vivo. Conflicts of interest The authors have no financial conflicts of interest. Acknowledgments This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology (NRF2012R1A1A1001645), and by 2011 Research Grant from Kangwon National University. References Antunes, L.C., Buckner, M.M., Auweter, S.D., Ferreira, R.B., Lolic, P., Finlay, B.B., 2010. Inhibition of Salmonella host cell invasion by dimethyl sulfide. Appl. Environ. Microbiol. 76, 5300. Bauer, C., Duewell, P., Mayer, C., Lehr, H.A., Fitzgerald, K.A., Dauer, M., Tschopp, J., Endres, S., Latz, E., Schnurr, M., 2010. Colitis induced in mice with dextran sulfate sodium (DSS) is mediated by the NLRP3 inflammasome. Gut 59, 1192. Bulua, A.C., Simon, A., Maddipati, R., Pelletier, M., Park, H., Kim, K.Y., Sack, M.N., Kastner, D.L., Siegel, R.M., 2011. Mitochondrial reactive oxygen species promote production of proinflammatory cytokines and are elevated in TNFR1-associated periodic syndrome (TRAPS). J. Exp. Med. 208, 519. Cassel, S.L., Eisenbarth, S.C., Iyer, S.S., Sadler, J.J., Colegio, O.R., Tephly, L.A., Carter, A.B., Rothman, P.B., Flavell, R.A., Sutterwala, F.S., 2008. The NALP3 inflammasome is essential for the development of silicosis. Proc. Natl. Acad. Sci. U. S. A. 105, 9035. Chang, C.K., Llanes, S., Schumer, W., 1999. Inhibitory effect of dimethyl sulfoxide on nuclear factor-kappa B activation and intercellular adhesion molecule 1 gene expression in septic rats. J. Surg. Res. 82, 294. Colucci, M., Maione, F., Bonito, M.C., Piscopo, A., Di Giannuario, A., Pieretti, S., 2008. New insights of dimethyl sulphoxide effects (DMSO) on experimental in vivo models of nociception and inflammation. Pharmacol. Res. 57, 419. Da Violante, G., Zerrouk, N., Richard, I., Provot, G., Chaumeil, J.C., Arnaud, P., 2002. Evaluation of the cytotoxicity effect of dimethyl sulfoxide (DMSO) on CaCO2 /TC7 colon tumor cell cultures. Biol. Pharm. Bull. 25, 1600. Dinarello, C., Arend, W., Sims, J., Smith, D., Blumberg, H., O’Neill, L., GoldbachMansky, R., Pizarro, T., Hoffman, H., Bufler, P., Nold, M., Ghezzi, P., Mantovani, A., Garlanda, C., Boraschi, D., Rubartelli, A., Netea, M., van der Meer, J., Joosten, L., Mandrup-Poulsen, T., Donath, M., Lewis, E., Pfeilschifter, J., Martin, M., Kracht, M., Muehl, H., Novick, D., Lukic, M., Conti, B., Solinger, A., Kelk, P., van de Veerdonk, F., Gabel, C., 2010. IL-1 family nomenclature. Nat. Immunol. 11, 973. Dostert, C., Petrilli, V., Van Bruggen, R., Steele, C., Mossman, B.T., Tschopp, J., 2008. Innate immune activation through NALP3 inflammasome sensing of asbestos and silica. Science 320, 674. Englen, M.D., Valdez, Y.E., Lehnert, N.M., Lehnert, B.E., 1995. Granulocyte/macrophage colony-stimulating factor is expressed and secreted in cultures of murine L929 cells. J. Immunol. Methods 184, 281. Hollebeeck, S., Raas, T., Piront, N., Schneider, Y.J., Toussaint, O., Larondelle, Y., During, A., 2011. Dimethyl sulfoxide (DMSO) attenuates the inflammatory response in the in vitro intestinal Caco-2 cell model. Toxicol. Lett. 206, 268. Hsu, H.Y., Twu, Y.C., 2000. Tumor necrosis factor-alpha -mediated protein kinases in regulation of scavenger receptor and foam cell formation on macrophage. J. Biol. Chem. 275, 41035. Jacob, S.W., Herschler, R., 1986. Pharmacology of DMSO. Cryobiology 23, 14. Kloesch, B., Liszt, M., Broell, J., Steiner, G., 2011. Dimethyl sulphoxide and dimethyl sulphone are potent inhibitors of IL-6 and IL-8 expression in the human chondrocyte cell line C-28/I2. Life Sci. 89, 473. Lamkanfi, M., 2011. Emerging inflammasome effector mechanisms. Nat. Rev. Immunol. 11, 213. Lamkanfi, M., Dixit, V.M., 2011. Modulation of inflammasome pathways by bacterial and viral pathogens. J. Immunol. 187, 597.

Lamkanfi, M., Dixit, V.M., 2012. Inflammasomes and their roles in health and disease. Annu. Rev. Cell Dev. Biol. 28, 137. Lee, C.A., Jones, B.D., Falkow, S., 1992. Identification of a Salmonella typhimurium invasion locus by selection for hyperinvasive mutants. Proc. Natl. Acad. Sci. U. S. A. 89, 1847. Lee, G.S., Subramanian, N., Kim, A.I., Aksentijevich, I., Goldbach-Mansky, R., Sacks, D.B., Germain, R.N., Kastner, D.L., Chae, J.J., 2012. The calcium-sensing receptor regulates the NLRP3 inflammasome through Ca2+ and cAMP. Nature 492, 123. Loppnow, H., Libby, P., 1990. Proliferating or interleukin 1-activated human vascular smooth muscle cells secrete copious interleukin 6. J. Clin. Invest. 85, 731. Mariathasan, S., Weiss, D.S., Newton, K., McBride, J., O’Rourke, K., Roose-Girma, M., Lee, W.P., Weinrauch, Y., Monack, D.M., Dixit, V.M., 2006. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature 440, 228. McDermott, M.F., Aksentijevich, I., Galon, J., McDermott, E.M., Ogunkolade, B.W., Centola, M., Mansfield, E., Gadina, M., Karenko, L., Pettersson, T., McCarthy, J., Frucht, D.M., Aringer, M., Torosyan, Y., Teppo, A.M., Wilson, M., Karaarslan, H.M., Wan, Y., Todd, I., Wood, G., Schlimgen, R., Kumarajeewa, T.R., Cooper, S.M., Vella, J.P., Amos, C.I., Mulley, J., Quane, K.A., Molloy, M.G., Ranki, A., Powell, R.J., Hitman, G.A., O’Shea, J.J., Kastner, D.L., 1999. Germline mutations in the extracellular domains of the 55 kDa TNF receptorTNFR1, define a family of dominantly inherited autoinflammatory syndromes. Cell 97, 133. Medzhitov, R., 2001. Toll-like receptors and innate immunity. Nat. Rev. Immunol. 1, 135. Mendez, C., Garcia, I., Maier, R.V., 1995. Antioxidants attenuate endotoxin-induced activation of alveolar macrophages. Surgery 118, 412. Murakami, T., Ockinger, J., Yu, J., Byles, V., McColl, A., Hofer, A.M., Horng, T., 2012. Critical role for calcium mobilization in activation of the NLRP3 inflammasome. Proc. Natl. Acad. Sci. U. S. A. 109, 11282. Nakahira, K., Haspel, J.A., Rathinam, V.A., Lee, S.J., Dolinay, T., Lam, H.C., Englert, J.A., Rabinovitch, M., Cernadas, M., Kim, H.P., Fitzgerald, K.A., Ryter, S.W., Choi, A.M., 2011. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat. Immunol. 12, 222. Phillis, J.W., Estevez, A.Y., O’Regan, M.H., 1998. Protective effects of the free radical scavengers, dimethyl sulfoxide and ethanol, in cerebral ischemia in gerbils. Neurosci. Lett. 244, 109. Rathinam, V.A., Vanaja, S.K., Fitzgerald, K.A., 2012. Regulation of inflammasome signaling. Nat. Immunol. 13, 333. Reimann, T., Buscher, D., Hipskind, R.A., Krautwald, S., Lohmann-Matthes, M.L., Baccarini, M., 1994. Lipopolysaccharide induces activation of the Raf-1/MAP kinase pathway. A putative role for Raf-1 in the induction of the IL-1 beta and the TNF-alpha genes. J. Immunol. 153, 5740. Rossol, M., Pierer, M., Raulien, N., Quandt, D., Meusch, U., Rothe, K., Schubert, K., Schoneberg, T., Schaefer, M., Krugel, U., Smajilovic, S., Brauner-Osborne, H., Baerwald, C., Wagner, U., 2012. Extracellular Ca2+ is a danger signal activating the NLRP3 inflammasome through G protein-coupled calcium sensing receptors. Nat. Commun. 3, 1329. Santos, N.C., Figueira-Coelho, J., Martins-Silva, J., Saldanha, C., 2003. Multidisciplinary utilization of dimethyl sulfoxide: pharmacological, cellular, and molecular aspects. Biochem. Pharmacol. 65, 1035. Schroder, K., Zhou, R., Tschopp, J., 2010. The NLRP3 inflammasome: a sensor for metabolic danger? Science 327, 296. Sun, J.S., Hahn, T.W., 2012. Comparative proteomic analysis of Salmonella enterica serovars enteritidis typhimurium and gallinarum. J. Vet. Med. Sci. 74, 285. van de Veerdonk, F.L., Smeekens, S.P., Joosten, L.A., Kullberg, B.J., Dinarello, C.A., van der Meer, J.W., Netea, M.G., 2010. Reactive oxygen species-independent activation of the IL-1beta inflammasome in cells from patients with chronic granulomatous disease. Proc. Natl. Acad. Sci. U. S. A. 107, 3030. van Lenten, B.J., Fogelman, A.M., 1992. Lipopolysaccharide-induced inhibition of scavenger receptor expression in human monocyte-macrophages is mediated through tumor necrosis factor-alpha. J. Immunol. 148, 112. Xaus, J., Comalada, M., Valledor, A.F., Lloberas, J., Lopez-Soriano, F., Argiles, J.M., Bogdan, C., Celada, A., 2000. LPS induces apoptosis in macrophages mostly through the autocrine production of TNF-alpha. Blood 95, 3823. Xing, L., Remick, D.G., 2005. Mechanisms of dimethyl sulfoxide augmentation of IL-1 beta production. J. Immunol. 174, 6195. Zhou, R., Yazdi, A.S., Menu, P., Tschopp, J., 2011. A role for mitochondria in NLRP3 inflammasome activation. Nature 469, 221. Zhu, W., Downey, J.S., Gu, J., Di Padova, F., Gram, H., Han, J., 2000. Regulation of TNF expression by multiple mitogen-activated protein kinase pathways. J. Immunol. 164, 6349.

Dimethyl sulfoxide inhibits NLRP3 inflammasome activation.

Dimethyl sulfoxide (DMSO) is an amphipathic molecule that is commonly/widely used as a solvent for biological compounds. In addition, DMSO has been st...
2MB Sizes 0 Downloads 0 Views