Immunology Letters 158 (2014) 143–150

Contents lists available at ScienceDirect

Immunology Letters journal homepage: www.elsevier.com/locate/immlet

Korean red ginseng extracts inhibit NLRP3 and AIM2 inflammasome activation Jeeyoung Kim a,1 , Huijeong Ahn a,1 , Byung-Cheol Han b , Seung-Ho Lee b , Young-Wook Cho c , Cheon Ho Kim a , Eui-Ju Hong e , Beum-Soo An d , Eui-Bae Jeung e , Geun-Shik Lee a,∗ a

College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 200-701, Republic of Korea Korea Ginseng Corp., Daejeon 305-805, Republic of Korea c Korean Basic Science Institute, Chuncheon 200-701, Republic of Korea d Department of Biomaterial Science, College of Natural Resources and Life Science, Pusan National University, Gyeongsangnam-do 627-706, Republic of Korea e Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungcheongbuk-do 361-763, Republic of Korea b

a r t i c l e

i n f o

Article history: Received 12 September 2013 Received in revised form 19 December 2013 Accepted 20 December 2013 Available online 10 January 2014 Keywords: Red ginseng extracts Cytokine Inflammasome Interleukin-1 Macrophages AIM2

a b s t r a c t Korean red ginseng extract (RGE) is one of the most popular natural herbs modulating the immune system. Although the effects of RGE on immunity have been reported, its effects on inflammasomes, multi-protein complexes that activate caspase-1 to induce maturation of interleukin (IL)-1␤, have not been studied yet. In this study, we elucidated the effect of RGE on inflammasome activation using mouse and human macrophages. In our results, RGE inhibited IL-1␤ maturation resulting from NLRP3 inflammasome activation in both in vitro and in vivo models. In addition, RGE strongly attenuated IL-1␤ secretion as well as pathogen clearance via pyroptotic cell death by macrophages through inhibition of AIM2 inflammasome activation. Ginsenosides Rg1 and Rh3 were suggested as inhibitors of the inflammasome activation. Thus, we demonstrated that RGE inhibits both NLRP3 and AIM2 inflammasome activation, with predominant involvement of the AIM2 inflammasome. Crown Copyright © 2014 Published by Elsevier B.V. All rights reserved.

1. Introduction Korean red ginseng (Panax Ginseng C.A Meyer) is one of the most well studied traditional medicinal herbs. Ginseng can be separated into red ginseng and white ginseng according to commercially use. Naturally dried ginseng is known as white ginseng, whereas red ginseng is made by steaming and drying fresh ginseng root. Korean red ginseng contains various active components, including saponins such as ginsenosides and non-saponins such as polysaccharides, peptides, fatty acids, and mineral oils [1]. Ginsenosides are the major active component of ginseng and has vasorelaxation, anti-oxidation, and anti-cancer effects [2]. Ginsenosides exhibit anti-inflammatory properties by inhibiting NF-␬B activation in association with reduced secretion and/or mRNA expression of proinflammatory mediators [3]. Ginsenosides are also amphiphilic in

∗ Corresponding author. Tel.: +82 33 250 8683; fax: +82 33 244 2367. E-mail address: [email protected] (G.-S. Lee). 1 These authors contributed equally to this work.

nature and have the ability to intercalate into the plasma membrane [4]. As ginsenosides are signaling molecules that are similar to steroid hormones, they can alter membrane fluidity and function as well as elicit cellular responses when moving through the plasma membrane [5,6]. Red ginseng has been studied for many years as an immune-modulating substance, and previous studies have determined it to be an adjuvant or remedy for the treatment of infectious diseases as well as many other metabolic disorders [2]. Interleukin (IL)-1␤ is an important mediator of inflammation that increases synthesis of cytokines, febrile disease progression, gene regulation of endometrial cells, chemotaxis, leukocyte adhesion, fibroblast activity, and heart rate [7]. IL-1␤ is generated by inflammasomes [8], which are cytoplasmic multi-protein complexes that mediate the maturation of IL-1␤ by activating caspase-1 [9]. The primary activity of inflammasomes is the activation of caspase-1. In detail, the caspase-1 catalyzes pro-IL-1␤ produced by PRRs signaling into mature IL-1␤ [10]. Most cytokines are activated as they are simultaneously generated and secreted, but IL-1␤ is first generated as a pro-form (pro-IL-1␤) before being converted into

0165-2478/$ – see front matter. Crown Copyright © 2014 Published by Elsevier B.V. All rights reserved. http://dx.doi.org/10.1016/j.imlet.2013.12.017

144

J. Kim et al. / Immunology Letters 158 (2014) 143–150

its final active form by inflammasome activation. Inflammasomes have been found to regulate other important aspects of inflammation and tissue repair such as pyroptosis, a form of cell death [11,12]. Mutation of just one inflammasome-associated gene has been shown to induce auto-inflammatory disease [13]. Several inflammasomes have been identified to date [10,14], including NLRP3 (NACHT, LRR, and PYD domains-containing protein 3) inflammasome, which recognizes various danger signals, AIM2 (absent in melanoma 2) inflammasome, which recognizes dsDNA, and NLRC4 (NLR Family, CARD domain-containing 4) inflammasome, which recognizes flagellin. Although the anti-inflammatory effects of Korean red ginseng extracts (RGE) have been widely reported, the effects of RGE on inflammasome activation have not been identified. In this study, we investigated the effects of RGE on IL-1␤ regulation and inflammasome activation in bone marrow-derived macrophages (BMDMs). We observed that RGE inhibited both NLRP3 and AIM2 inflammasome activation. 2. Materials and methods 2.1. Cell culturing Unless otherwise indicated, all materials for cell culture were obtained from PAA (GE Healthcare Bio-Sciences Co., NJ, USA). Bone marrow-derived macrophages (BMDMs) were obtained by differentiating bone marrow progenitors from tibia and femur bones using L929 cell-conditioned medium (LCCM) as a source of granulocyte/macrophage colony-stimulating factor [15]. Progenitors were cultured in RPMI 1640 (LM 011-01, Welgene, Seoul, Republic of Korea) 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. THP-1 cells were obtained from Korean Cell Line Bank (KCLB No. 40202; Seoul, Republic of Korea) and maintained in RPMI 1640 medium containing 10% FBS, 100 U/ml of penicillin, and 100 ␮g/ml of streptomycin at 37 ◦ C in a 5% CO2 atmosphere. THP-1 cells were plated in 12-well plates (1 × 106 cells per well) and differentiated to macrophage-like cells with phorbol 12-myristate acetate (100 nM, PMA; Invivogen, CA, USA) for 72 h. 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 RGE for 1 h and the cytotoxicity was measured by Cell Counting Kit-8 (Dojindo Molecular Technologies, Inc., MD, USA) as manufacture’s protocol. 2.2. Korean red ginseng extract (RGE) Korean RGE was manufactured from the roots of a 6-yearold fresh Panax ginseng and provided by Korea Ginseng Corp. (Daejeon, Republic of Korea). The provided RGE was composed 33.75% of water, 5.5% of crude ash, 12.96% of crude protein, and 47.79% of carbohydrate. In addition, RGE contained Rb1 (9.29 mg/g), Rb2 (3.18 mg/g), Rc (3.60 mg/g), Rd (1.00 mg/g), Re (2.45 mg/g), Rf (1.06 mg/g), Rg1 (2.00 mg/g), Rg2 (0.90 mg/g), Rg3 (1.90 mg/g), Rh1 (0.65 mg/g) and other minor ginsenosides. 2.3. Inflammasome activation and inhibition The PMA treated THP-1 and BMDMs (1.0 × 106 cells per well) were plated on 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 or THP-1 cells were subjected to the following

activation steps for 1 or 3 h. For NLRP3 inflammasome activation, the medium was replaced with RPMI 1640 supplemented with ATP (2 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., Gyeonggido, Republic of Korea) for 3 h. For NLRC4 inflammasome activation, the medium was exchanged with Salmonella typhimurium (OD600: 1.2, 0.5%) 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, CA, USA) for 1 h or Listeria monocytogenes (OD600: 0.3, 10%) for 3 h. In addition, E. coli (DH5␣, OD600: 1.0, 1%) was used as a negative control. RGE, Quillaja saponin (Junsei Chemical Co., Tokyo, Japan), ginsenoside Rh1 (ChromaDex® Co., CA, USA), or ginsenoside Rg3 (ChromaDex® Co.) were co-treated with the above activators. Due to the cytotoxicity of saponin, Rh1, and Rg3 on BMDMs (data not shown), the optimized concentrations as indicated in the figure were adopted in the current experiment. Cellular supernatant (Sup), lysate (Lys), and cross-linked pellets (Pellet) with suberic acid bis (Sigma–Aldrich Co.) were collected for further analysis.

2.4. 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-human IL-1␤ antibody (AF-201-NA, R&D systems), anticaspase-1 p20 antibody (06-503, EMD Millipore Co, MA, USA), anti-caspase-1 antibody (sc-622, Santa Cruz Biotechnology), antiAsc antibody (sc-22514, Santa Cruz Biotechnology, CA, USA), c-Myc antibody (SC-40, Santa Cruz Biotechnology), or anti-actin antibody (sc-1615, Santa Cruz Biotechnology). The membranes were further probed with HRP-conjugated 2nd anti-sera (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).

2.5. IL-1ˇ detection using ELISA To quantitate the secreted IL-1␤, the cell culture supernatants of BMDMs were measured by mouse IL-1beta/IL-1F2 Quantikine ELISA Kit (R&D Systems). The ELISA plates were readout using a microplate reader (Bio-Red, CA, USA).

2.6. Animals Male C57BL/6 mice (8-week-old) were purchased from Narabio Co. (Seoul, Republic of Korea). All mice were maintained under a 12 h light/dark cycle at 24 ◦ C. Animals were provided standard sterile food and water ad libitum, after which they were allowed to adjust to the environment for 1 week. For LPS-induced endotoxin shock, mice were fed 2% of RGE (n = 11), 10% of RGE (n = 10) or water (n = 21) ad libitum for 1 week before LPS (20 mg/kg) intraperitoneal injection. Animals were observed every 8 h for 3 days. Animal care and all experimental procedures were conducted 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).

J. Kim et al. / Immunology Letters 158 (2014) 143–150

2.7. Transfection efficiency assay Transfection was carried out using LipofectamineTM 2000 (Invitrogen) like dsDNA-mediated AIM2 inflammasome activation. 293 T cells (3 × 105 cells) were plated on 6-well plates (SPL Lifesciences) 1 day before transfection. Plasmid (2 ␮g) expressing both c-Myc and red fluorescence protein (RFP) was mixed with serum-free DMEM containing LipofectamineTM 2000 (Invitrogen), after which the mixture was added to wells. After incubation for 1 h, cell culture media were replaced with DMEM containing 10% FBS with antibiotics for 17 h. RFP signals were observed with a fluorescence microscope (Nikon, Tokyo, Japan) and cellular lysates were subjected to Western blot assay.

145

inflammasome activation (Fig. 1D). The inhibitory properties of RGE on NLRP3 inflammasome were further confirmed in human macrophages (THP-1 cells, Fig. 1F). The inhibitory effect of RGE on NLRP3 inflammasome activation was further confirmed using an LPS-induced endotoxin shock model. According to the literature, susceptibility to LPS-induced endotoxin shock is reduced in NLRP3 gene-deleted mice compared to intact mice [17]. As shown in Fig. 1G, 2 or 10% of RGE orally treated mice showed higher survival rates than control mice 1 week prior to LPS injection. Thus, RGE attenuated the severity of NLRP3 inflammasome-mediated disease symptoms in animals, which is similar to our in vitro result in which RGE inhibited IL-1␤ maturation in BMDMs. Hence, RGE directly inhibited NLRP3 inflammasome activation and could be applied as an NLRP3 inflammasome inhibitor.

2.8. Bacterial growth 3.2. Korean red ginseng inhibits AIM2 inflammasome activation S. typhimurium and L. monocytogenes were obtained from the Korean Culture Center of Microorganisms (KCCM; Seoul, Republic of Korea). Salmonella and Listeria were grown on Luria-Bertani (LB, Laboratories Conda, Madrid, Spain) and Brain Heart Infusion (BHI, Laboratories Conda) agar plates for 18 h at 37 ◦ C. A single colony was transferred to LB or BHI broth, after which the culture was incubated for 18 h with shaking at 37 ◦ C. Listeria host invasion assay was performed as previously described [16]. Briefly, BMDMs (1 × 106 cells per well or 5 × 104 cells per well) were plated on 96-well plates and inoculated with Listeria (2 × 107 colony-forming units [cfu] or 1 × 105 cfu) in DMEM containing 10% FBS for 1 or 12 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 Listeria, followed by plating on a BHI plate to calculate cfu. 2.9. Statistical analyses Statistical analyses were performed using 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). 3. Results 3.1. Korean red ginseng inhibits NLRP3 inflammasome activation To elucidate the effect of red ginseng extracts (RGE) on inflammasome activation, we treated LPS-primed BMDMs with various dosages of RGE alone or RGE with ATP, a well-characterized NLRP3 inflammasome activator, and measured IL-1␤ (p17, active form) secretion by immuno blotting (Fig. 1A) and ELISA assays (Fig. 1B). As shown in Fig. 1A (left side), mature IL-1␤ (p17) was not detected in the cell supernatant (Sup) of the RGE alone treated cells suggesting that RGE did not activate inflammasomes. While we treated LPS-primed BMDMs with RGE in the presence of ATP to estimate the inhibitory effect of RGE on IL-1␤ secretion resulting from NLRP3 inflammasome activation. RGE dose-dependently inhibited ATP-mediated IL-1␤ secretion. In addition, RGE effectively interrupted the formation of Asc pyroptosomes, which are a typical indicator of inflammasome activation. As expected, the RGE treatment did not induce any cytotoxicity in the BMDMs (Fig. 1C). Further, we treated LPS-primed BMDMs with other NLRP3 inflammasome activators, including nigericin (NG; Fig. 1D) and aluminum potassium sulfate (Alum; Fig. 1E) in the presence of RGE to confirm inhibition of NLRP3 inflammasome activation. Whereas production of mature IL-1␤ was observed in macrophages treated with NG or Alum alone, RGE dose-dependently inhibited NLRP3 inflammasome activation. In addition, RGE attenuated the secretion of the active form of caspase-1 (p20) by NG-mediated NLRP3

We further tested the inhibitory effect of RGE on NLRC4 and AIM2 inflammasome activation. The NLRC4 inflammasome is activated by intracellular bacteria containing flagellin, whereas the AIM2 inflammasome activation is triggered by intracellular dsDNA derived from the host or pathogens [18]. As shown in Fig. 2A, we inoculated S. typhimurium (Salmonella) for NLRC4 inflammasome activation or transfected dsDNA into LPS-primed BMDMs for AIM2 inflammasome activation, after which IL-1␤ maturation and Asc pyroptosome formation were analyzed. Although RGE did not block IL-1␤ maturation or Asc pyrotosome formation induced by Salmonella, it strongly interrupted dsDNA-mediated IL-1␤ secretion and Asc pyrotosome formation. Interestingly, less than 600 ppm of RGE completely attenuated IL-1␤ (Fig. 2B) and caspase-1 (Fig. 2C) secretions. Thus, RGE could be an effective AIM2 inflammasome inhibitor. We further explored the mechanism of AIM2 inflammasome induction. To confirm whether or not RGE inhibited dsDNA transfection, a plasmid expressing Myc and RFP proteins was transfected into 293 T cells in the presence of various dosages of RGE (Fig. 2D). Although the highest dosage of RGE did reduce transfection efficiency, all other RGE concentrations that inhibited AIM2 inflammasome activation had no effect on Myc and RFP expressions. In addition, we inoculated L. monocytogenes (Listeria), which triggers both AIM2 and NLRP3 inflammasome activation with predominant involvement of the AIM2 inflammasome [16], into LPS-primed BMDMs in the presence of RGE (Fig. 2E). RGE significantly inhibited Listeria-induced IL-1␤ secretion, although the degree of inhibition was less potent than that of dsDNA-mediated IL-1␤ secretion. In the same vein as the transfection efficiency test (Fig. 2D), we inoculated Listeria into BMDMs and measured intracellular bacteria number (cfu) after 1 or 12 h of inoculation. As shown in Fig. 2F, the invasion rate of Listeria into BMDMs was not altered by the presence of RGE, indicating that RGE directly inhibited intracellular inflammasome activation. In addition, the number of Listeria after 12 h of inoculation significantly increased in the presence of RGE, implying that RGE interrupted clearance of Listeria via pyroptotic cell death [19] (Fig. 2G). The inhibitory effects of RGE on AIM2 inflammasome were further confirmed in human macrophages (Fig. 2H). Thus, RGE was a potent inhibitor on AIM2 inflammasome activation. 3.3. Ginsenosides inhibit inflammasome activation We next attempted to identify which material of red ginseng is responsible for the inhibition of inflammasome activation. We firstly focused on saponins (Quillaja saponins) and ginsenosides (Rh1 and Rg3; Fig. 3A), which are the major pharmacological constituents of ginseng [2]. We treated LPS-primed BMDMs with saponin, Rh1, or Rg3 along with the inflammasome activators NG or dsDNA. As shown in Fig. 3B, IL-1␤ secretion

146

J. Kim et al. / Immunology Letters 158 (2014) 143–150

Fig. 1. RGE inhibits NLRP3 inflammasome activation. BMDMs (1 × 106 cells per well) were primed by LPS (10 ␮g/ml) in RPMI medium containing 10% FBS for 3 h, after which the medium was replaced with RPMI medium containing the indicated percentages of RGE with ATP (2 mM, A and B) or NG (40 ␮M, D) for 1 h or Alum (200 ␮g/ml, E) for 3 h. Cell culture supernatants (Sup), cell lysates (Lys), and cross-linked pellets (Pellet) from whole-cell lysates were analyzed by immunoblotting as indicated. Secreted IL-1␤ was quantitated by an ELISA based assay kit and presented with bar graphs (B, D, and E). (C) The cytotoxicity of RGE was measured after applying the indicated dosage of RGE on BMDMs for 1 h like to the inflammasome activating step. Triton ×-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%. (F) THP-1 (1 × 106 cells per well) were differentiated by PMA (100 nM) for 3 days and then primed by LPS (10 ␮g/ml) for 3 h, after which the medium was replaced with RPMI medium containing the indicated percentages of RGE with ATP (2 mM) or NG (40 ␮M) for 1 h or Alum (200 ␮g/ml) for 3 h. Supernatants and cell lysates were analyzed by immunoblotting as indicated. All immunoblot data shown are representative of three or more independent experiments. All data for bar graphs represent the mean ± s.e.m. of three independent experiments, each performed in triplicate. (G) Mice were treated with 2 or 10% of RGE or water ad libitum for 1 week before LPS (20 mg/kg) intraperitoneal injection. Survival rates were observed for 3 days. P < 0.05, RGE (2 and 10%) treated mice vs. Water supplied mice.

was unaltered by saponin. However, the highest concentration of Rh1 moderately inhibited NG or dsDNA-induced IL-1␤ secretion (Fig. 3C). In addition, Rg3 ginsenoside attenuated inflammasome activator-mediated IL-1␤ maturation (Fig. 3D). Interestingly, Rg3 was the most potent inhibitor of dsDNAmediated AIM2 inflammasome activation. Thus the ginsenosides derived from RGE interrupted NLRP3 and AIM2 inflammasome activation. 4. Discussion The anti-inflammatory properties of RGE have been previously studied [20], although there is currently no study on the

effect of RGE on inflammasome activation mediating IL-1␤ maturation. In this study, we observed that RGE inhibited two well characterized inflammasomes, NLRP3 and AIM2. That is, RGE inhibited IL-1␤ secretion from NLRP3 activator-treated BMDMs and/or THP-1, and it ameliorated LPS-induced lethality. In addition, RGE strongly attenuated IL-1␤ secretion and pyroptosome formation in dsDNA-transfected macrophages. Attenuation of pyrototic cell death induced intracellular Listeria growth. As expected, ginsenosides Rh1 and Rg3 were shown to be key inhibitors of inflammasome activation, whereas saponins derived from quillaja did not alter inflammasome activation. Thus, ginsenosides of RGE act as inhibitors of both NLRP3 and AIM2 inflammasome activation.

J. Kim et al. / Immunology Letters 158 (2014) 143–150

147

Fig. 2. Effects of RGE on dsDNA- or Listeria-mediated AIM2 inflammasome activation. (A) LPS-primed BMDMs were co-treated with NLRC4 or AIM2 inflammasome activators, Salmonella (OD600: 1.2, 0.5%), or dsDNA (2 ␮g/ml) in Lipofectamine (4 ␮l/ml) as well as the indicated percentages of RGE. Cell culture supernatants (Sup), cell lysates (Lys), and cross-linked pellets (Pellet) from whole-cell lysates were analyzed by immunoblotting as indicated. The secreted IL-1␤ was quantitated with an ELISA-based assay and presented as bar graphs. Data represent the mean ± s.e.m. of three independent experiments. (B) The inhibitory property of RGE on dsDNA-mediated IL-1␤ secretion was progressively observed in the present of serial dilution of RGE. (C) Caspase-1 maturation and secretion was detected by Western blotting assay. (D) Plasmid expressing Myc and red fluorescence protein (RFP) was introduced into 293 T cells with Lipofectamine 2000 in the presence of RGE. Red fluorescence signals were observed with fluorescence microscope. Bar indicated 100 ␮m. Myc band intensity was measured to elucidate the effect of RGE on transfection efficiency. Myc expression without RGE was set as 100%, and the non-transfected sample was set as 0%. Data represent the mean ± s.e.m. of three independent experiments. (E) LPS-primed BMDMs were inoculated with the indicated bacteria with or without RGE and measured for IL-1␤ secretion. BMDMs were inoculated with Listeria for 1 h (F) or 12 h (G), after which the number of intracellular bacteria was calculated. cfu of the Listeria-inoculated group was set as 100%, and data represent the mean ± s.e.m. of four independent experiments, each performed in triplicate. (H) Human macrophages (THP-1) were primed by LPS and then treated Salmonella, dsDNA, or Listeria in the present of RGE. Supernatants and cell lysates were analyzed by immunoblotting as indicated.

148

J. Kim et al. / Immunology Letters 158 (2014) 143–150

Fig. 3. Effects of ginsenosides on NLRP3 or AIM2 inflammasome activation. (A) The chemical structures of ginsenosides, Rh1 and Rg3, were adopted from PubChem (http://pubchem.ncbi.nlm.nih.gov). LPS-primed BMDMs were treated with NG (40 ␮M) or dsDNA (2 ␮g/ml) in Lipofectamine (4 ␮l/ml) in the presence of saponins (Quillaja saponins, (B) ginsenoside Rh1, (C) or ginsenoside Rg3 (D). Cell culture supernatants (Sup) and cell lysates (Lys) were analyzed by immunoblotting as indicated.

Although several inflammasomes have been identified [9], their molecular mechanisms of activation have been poorly characterized, except for the NLRP3 inflammasome. Briefly, inflammasome activation might be controlled by intracellular potassium efflux, mitochondrial reactive oxygen species (ROS) generation, and

lysosomal rupture [18,21]. In addition, intracellular Ca2+ and cAMP have been suggested as alternative molecular effectors in the regulation of NLRP3 inflammasome activation [22]. Among these, we have focused on mitochondrial ROS [23–25] and intracellular Ca2+ [22,26,27] as mediators of the inhibitory effect of RGE on NLRP3

J. Kim et al. / Immunology Letters 158 (2014) 143–150

inflammasome, as they have been frequently reported as targets of RGE. That is, ginseng extract has been shown to reverse elevation of ROS generation [28–30] and intracellular Ca2+ [31–33] in various cell types. Based on this evidence, we conclude that RGE inhibits NLRP3 inflammasome activation. To elucidate the inhibitory effect of RGE on inflammasome activation in an animal model, we performed an LPS lethality test (Fig. 1G). As expected, RGE-fed mice showed higher survival rates than control mice. Similar data have previously been reported [34]. In a study by Ahn et al., pretreatment with ginsan, a polysaccharide extracted from P. ginseng, protected mice from lethality induced by septic challenge, and ginsan-treated mice displayed lower levels of serum pro-inflammatory cytokines such as IL-1␤ [34]. In addition, ginsan reduced the expression of toll-like receptor (TLR)-2, -4, -9, and MyD88, thereby protecting mice from sepsis [34]. Based on this report, we speculate that RGE inhibits both IL-1␤ transcription and maturation via inflammasome activation. RGE not only inhibited NLRP3 inflammasome activation but also strongly attenuated dsDNA-induced IL-1␤ secretion. Cytoplasmic dsDNA sensed by AIM2 interacts with Asc, resulting in the formation of the Asc pyroptosome for induction of pyroptotic cell death in cells containing caspase-1 [18,35]. Pyroptosis is a critical pro-inflammatory cell death mechanism involving caspase-1 activity in which inflammasomes mediate pathogen clearance [9]. To confirm the inhibitory effect of RGE on Asc pyroptosome formation, we used L. monocytogenes, which displays predominantly AIM2 inflammasome activation over NLRP3 inflammasome activation [16]. The invasion rate of Listeria was unaffected by RGE treatment, whereas the growth rate of Listeria was significantly elevated. This result suggests that RGE inhibits both AIM2 and NLRP3 inflammasome activation and interrupts clearance of Listeria. Ginsenosides can be divided into protpanaxadiol (PPD) and protophanaxatriol (PPT) ginsenosides according to their basic structure [2]. PPD type ginsenosides include ginsenosides-Rb1, Rb2, -Rc, -Rd, -Rg3, and -Rh, whereas PPT type ginsenosides include ginsenosides-Re, -Rf, -Rg1, -Rg2 and -Rh1 [2]. In the current study, we selected two ginsenosides, Rg3 or Rh1, and saponins (Quillaja saponins) and determined their effects on inflammasome activation. Rh1, which possesses anti-allergic and anti-inflammatory activities, has been recently reported to ameliorate obesity and myocardial injury in the rodent models [36–38]. While Rg3 has been progressive studied as an anti-carcinogenic agent and it also presents several biological activities including anti-oxidant, antiangiogenesis, and anti-inflammatory [39,40]. Rg3 is found mainly in red ginseng and is derived from the hydrolysis of saponins by heat processing [2]. Although both Rh1 and Rg3 have showed anti-inflammatory activities, the effect of these on inflammasome activation has not been studied. As shown in Fig. 3, saponins did not inhibit NLRP3 or AIM2 inflammasome activation however Rh1 and Rg3 attenuated NG- or dsDNA-induced IL-1␤ secretion. Especially, Rg3 seemed to more potently inhibit dsDNA-induced IL-1␤ secretion. Other studies have proposed that various metabolic diseases are associated with inflammasome activation [41]. Indeed, treatments targeting IL-1␤ using an IL-1 receptor antagonist or IL-1␤ monoclonal antibody have been suggested to cure arthritis, type 2 diabetes, arteriosclerosis, or Alzheimer’s disease [42–44]. However, these targeted treatments impose large financial burdens as well as unwanted side effects on patients. Hence, selective inflammasome inhibitors are needed such as glyburide, a type 2 diabetes drug, and the NF-␬B inhibitors Parthenolide and Bay11-7082, which were reported as selective inhibitors of NLRP3 inflammasome activation [45,46]. In the current study, we suggest RGE as an inhibitor of inflammasome activation or an alternative source for IL-1␤targeted treatments.

149

Conflict of interest The authors declare no financial or commercial conflict 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 (NRF-2012R1A1A1001645), by a grant from the Next-Generation BioGreen 21 Program (No. PJ009069), Rural Development Administration, Republic of Korea, and by a 2011 Research Grant from Kangwon National University. References [1] Baek SH, Bae ON, Park JH. Recent methodology in ginseng analysis. Journal of Ginseng Research 2012;36:119–34. [2] Kang S, Min H. Ginseng, the ‘Immunity Boost’: the effects of Panax ginseng on immune system. Journal of Ginseng Research 2012;36:354–68. [3] Lu JM, Yao Q, Chen C. Ginseng compounds: an update on their molecular mechanisms and medical applications. Current Vascular Pharmacology 2009;7:293–302. [4] Attele AS, Wu JA, Yuan CS. Ginseng pharmacology: multiple constituents and multiple actions. Biochemical Pharmacology 1999;58:1685–93. [5] Lee YJ, Chung E, Lee KY, Lee YH, Huh B, Lee SK. Ginsenoside-Rg1, one of the major active molecules from Panax ginseng, is a functional ligand of glucocorticoid receptor. Molecular and Cellular Endocrinology 1997;133:135–40. [6] Lee Y, Jin Y, Lim W, Ji S, Choi S, Jang S, et al. A ginsenoside-Rh1, a component of ginseng saponin, activates estrogen receptor in human breast carcinoma MCF-7 cells. Journal of Steroid Biochemistry and Molecular Biology 2003;84:463–8. [7] Dinarello CA. Biologic basis for interleukin-1 in disease. Blood 1996;87:2095–147. [8] Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Molecular Cell 2002;10:417–26. [9] Lamkanfi M, Dixit VM. Inflammasomes and their roles in health and disease. Annual Review of Cell and Developmental Biology 2012;28:137–61. [10] Schroder K, Tschopp J. The inflammasomes. Cell 2010;140:821–32. [11] Lamkanfi M, Dixit VM. Modulation of inflammasome pathways by bacterial and viral pathogens. Journal of Immunology 2011;187:597–602. [12] Lamkanfi M. Emerging inflammasome effector mechanisms. Nature Reviews Immunology 2011;11:213–20. [13] McDermott MF, Aksentijevich I, Galon J, McDermott EM, Ogunkolade BW, Centola M, et al. Germline mutations in the extracellular domains of the 55 kDa TNF receptor, TNFR1, define a family of dominantly inherited autoinflammatory syndromes. Cell 1999;97:133–44. [14] Schroder K, Zhou R, Tschopp J. The NLRP3 inflammasome: a sensor for metabolic danger. Science 2010;327:296–300. [15] Englen MD, Valdez YE, Lehnert NM, Lehnert BE. Granulocyte/macrophage colony-stimulating factor is expressed and secreted in cultures of murine L929 cells. Journal of Immunological Methods 1995;184:281–3. [16] Kim S, Bauernfeind F, Ablasser A, Hartmann G, Fitzgerald KA, Latz E, et al. Listeria monocytogenes is sensed by the NLRP3 and AIM2 inflammasome. European Journal of Immunology 2010;40:1545–51. [17] Mariathasan S, Weiss DS, Newton K, McBride J, O’Rourke K, Roose-Girma M, et al. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature 2006;440:228–32. [18] Hornung V, Ablasser A, Charrel-Dennis M, Bauernfeind F, Horvath G, Caffrey DR, et al. AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature 2009;458:514–8. [19] Sauer JD, Witte CE, Zemansky J, Hanson B, Lauer P, Portnoy DA. Listeria monocytogenes triggers AIM2-mediated pyroptosis upon infrequent bacteriolysis in the macrophage cytosol. Cell Host & Microbe 2010;7:412–9. [20] Hofseth LJ, Wargovich MJ. Inflammation, cancer, and targets of ginseng. Journal of Nutrition 2007;137:183S–5S. [21] Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nature Immunology 2010;11:136–40. [22] Lee GS, Subramanian N, Kim AI, Aksentijevich I, Goldbach-Mansky R, Sacks DB, et al. The calcium-sensing receptor regulates the NLRP3 inflammasome through Ca2+ and cAMP. Nature 2012;492:123–7. [23] Martinon F. Signaling by ROS drives inflammasome activation. European Journal of Immunology 2010;40:616–9. [24] Sorbara MT, Girardin SE. Mitochondrial ROS fuel the inflammasome. Cell Research 2011;21:558–60. [25] Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature 2011;469:221–5. [26] Murakami T, Ockinger J, Yu J, Byles V, McColl A, Hofer AM, et al. Critical role for calcium mobilization in activation of the NLRP3 inflammasome.

150

[27]

[28]

[29]

[30]

[31]

[32]

[33]

[34]

[35]

J. Kim et al. / Immunology Letters 158 (2014) 143–150 Proceedings of the National Academy of Sciences of the United States of America 2012;109:11282–7. Rossol M, Pierer M, Raulien N, Quandt D, Meusch U, Rothe K, et al. Extracellular Ca2+ is a danger signal activating the NLRP3 inflammasome through G proteincoupled calcium sensing receptors. Nature Communications 2012;3:1329. Im GJ, Chang JW, Choi J, Chae SW, Ko EJ, Jung HH. Protective effect of Korean red ginseng extract on cisplatin ototoxicity in HEI-OC1 auditory cells. Phytotherapy Research: PTR 2010;24:614–21. Hwang HJ, Kwak YS, Yoon GA, Kang MH, Park JH, Lee BK, et al. Combined effects of swim training and ginseng supplementation on exercise performance time, ROS, lymphocyte proliferation, and DNA damage following exhaustive exercise stress. International Journal for Vitamin and Nutrition Research/Internationale Zeitschrift fur Vitamin – und Ernahrungsforschung/Journal international de vitaminologie et de nutrition 2007;77:289–96. Lin E, Wang Y, Mehendale S, Sun S, Wang CZ, Xie JT, et al. Antioxidant protection by American ginseng in pancreatic beta-cells. American Journal of Chinese Medicine 2008;36:981–8. Kim YC, Kim SR, Markelonis GJ, Oh TH. Ginsenosides Rb1 and Rg3 protect cultured rat cortical cells from glutamate-induced neurodegeneration. Journal of Neuroscience Research 1998;53:426–32. Kim SJ, Jeong HJ, Yi BJ, Kang TH, An NH, Lee EH, et al. Transgenic Panax ginseng inhibits the production of TNF-alpha, IL-6, and IL-8 as well as COX-2 expression in human mast cells. American Journal of Chinese Medicine 2007;35:329–39. Rhim H, Kim H, Lee DY, Oh TH, Nah SY. Ginseng and ginsenoside Rg3, a newly identified active ingredient of ginseng, modulate Ca2+ channel currents in rat sensory neurons. European Journal of Pharmacology 2002;436:151–8. Ahn JY, Choi IS, Shim JY, Yun EK, Yun YS, Jeong G, et al. The immunomodulator ginsan induces resistance to experimental sepsis by inhibiting Toll-like receptor-mediated inflammatory signals. European Journal of Immunology 2006;36:37–45. Fernandes-Alnemri T, Yu JW, Datta P, Wu J, Alnemri ES. AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature 2009;458:509–13.

[36] Gu W, Kim KA, Kim DH, Ginsenoside. Rh1 ameliorates high fat diet-induced obesity in mice by inhibiting adipocyte differentiation. Biological & Pharmaceutical Bulletin 2013;36:102–7. [37] Gai Y, Ma Z, Yu X, Qu S, Sui D. Effect of ginsenoside Rh1 on myocardial injury and heart function in isoproterenol-induced cardiotoxicity in rats. Toxicology Mechanisms and Methods 2012;22:584–91. [38] Park EK, Choo MK, Han MJ, Kim DH. Ginsenoside Rh1 possesses antiallergic and anti-inflammatory activities. International Archives of Allergy and Immunology 2004;133:113–20. [39] Sun C, Gao W, Zhao B, Cheng L. Optimization of the selective preparation of 20(R)-ginsenoside Rg3 catalyzed by d,l-tartaric acid using response surface methodology. Fitoterapia 2013;84:213–21. [40] Shin YM, Jung HJ, Choi WY, Lim CJ. Antioxidative, anti-inflammatory, and matrix metalloproteinase inhibitory activities of 20(S)-ginsenoside Rg3 in cultured mammalian cell lines. Molecular Biology Reports 2013;40: 269–79. [41] Wen H, Ting JP, O’Neill LA. A role for the NLRP3 inflammasome in metabolic diseases – did Warburg miss inflammation. Nature Immunology 2012;13:352–7. [42] Aksentijevich I, Masters SL, Ferguson PJ, Dancey P, Frenkel J, van Royen-Kerkhoff A, et al. An autoinflammatory disease with deficiency of the interleukin-1-receptor antagonist. New England Journal of Medicine 2009;360:2426–37. [43] Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature 2013;493:674–8. [44] Grant RW, Dixit VD. Mechanisms of disease: inflammasome activation and the development of type 2 diabetes. Frontiers in Immunology 2013;4:50. [45] Juliana C, Fernandes-Alnemri T, Wu J, Datta P, Solorzano L, Yu JW, et al. Antiinflammatory compounds parthenolide and Bay 11-7082 are direct inhibitors of the inflammasome. Journal of Biological Chemistry 2010;285:9792–802. [46] Lamkanfi M, Mueller JL, Vitari AC, Misaghi S, Fedorova A, Deshayes K, et al. Glyburide inhibits the Cryopyrin/Nalp3 inflammasome. Journal of Cell Biology 2009;187:61–70.

Korean red ginseng extracts inhibit NLRP3 and AIM2 inflammasome activation.

Korean red ginseng extract (RGE) is one of the most popular natural herbs modulating the immune system. Although the effects of RGE on immunity have b...
2MB Sizes 4 Downloads 0 Views