Ginger and Zingerone Ameliorate Lipopolysaccharide-Induced Acute Systemic Inflammation in Mice, Assessed by Nuclear Factor-κB Bioluminescent Imaging Chien-Yun Hsiang,a,1 Hui-Man Cheng,b,1 Hsin-Yi Lo,c Chia-Cheng Li,d Pei-Chi Chou,b Yu-Chen Lee,e Tin-Yun Ho*,c,g a Department of Microbiology, China Medical University, Taichung 40402, Taiwan b c School of Chinese Medicine, China Medical University, Taichung 40402, Taiwan Graduate Institute of Chinese Medicine, China Medical University, Taichung 40402, Taiwan d Graduate Institute of Cancer Biology, China Medical University, Taichung 40402, Taiwan e Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan g Department of Health and Nutrition Biotechnology, Asia University, Taichung 41354, Taiwan *Corresponding author. Telephone: +886 4 22053366 ext. 3302. Fax: +886 4 22032295. E-mail: cyhsiang@mail.cmu.edu.tw 1 These authors equally contributed to this work. Short title: ginger and zingerone ameliorate LPS-induced inflammation 1 1 ABSTRACT 2 Ginger is a commonly used spice in cooking. In this study, we comprehensively evaluated 3 the anti-inflammatory activities of ginger and its component zingerone in lipopolysaccharide 4 (LPS)-induced acute systemic inflammation in mice via nuclear factor-κB (NF-κB) 5 bioluminescent imaging. Ginger and zingerone significantly suppressed LPS-induced NF-κB 6 activities in cells in a dose-dependent manner, and the maximal inhibition (84.5±3.5% and 7 96.2±0.6%) was observed at 100 μg/ml ginger and zingerone, respectively. Moreover, dietary 8 ginger and zingerone significantly reduced LPS-induced proinflammatory cytokine 9 production in sera by 62.9±18.2% and 81.3±6.2%, respectively, and NF-κB bioluminescent 10 signals in whole body by 26.9±14.3% and 38.5±6.2%, respectively. In addition, ginger and 11 zingerone suppressed LPS-induced NF-κB-driven luminescent intensities in most organs and 12 the maximal inhibition by ginger and zingerone was observed in small intestine. 13 Immunohistochemical staining further showed that ginger and zingerone decreased 14 interleukin-1β (IL-1β)-, CD11b-, and p65-positive areas in jejunum. In conclusion, our 15 findings suggested that ginger and zingerone were likely to be broad-spectrum 16 anti-inflammatory agents in most organs that suppressed the activation of NF-κB, the 17 production of IL-1β, and the infiltration of inflammatory cells in mice. 18 19 Key words: Ginger, zingerone, nuclear factor-κB, inflammation, bioluminescent imaging 2 20 INTRODUCTION 21 Ginger, the rhizome of Zingiber officinale, is one of the most commonly used spices in 22 cooking. It is also a frequently used herb in alternative medicines. Clinical studies have 23 shown that ginger is effective in ameliorating nausea and vomiting caused by anti-retroviral 24 therapy, pregnancy, post-operation, and chemotherapy.1,2 It has add-on effects on reducing 25 knee pain and improving knee function in patients with symptomatic knee osteoarthritis.3,4 It 26 is effective on pain relief in primary dysmenorrhea, eccentric exercise, and migraine.5,6 27 Moreover, consumption of ginger is useful for patients with type 2 diabetes due to the 28 reduction of glycated hemoglobin and the improvement of insulin resistance.7 These clinical 29 data indicate the pharmacological application of ginger in medicine. 30 Anti-inflammatory activities of ginger and its ingredients have been suggested in in vitro 31 studies. For example, ginger extract inhibits the production of nitric oxide (NO) and 32 proinflammatory cytokines in lipopolysaccharide (LPS)-stimulated microglial cells, inhibits 33 the activation of macrophages, and reduces the production of LPS-induced proinflammatory 34 chemokines in bronchial epithelial cells.8,9 Moreover, ginger constituents, such as 6-shogaol, 35 gingerol and 6-dehydroginerdione, display anti-inflammatory potentials in LPS-induced 36 microglial cells or macrophages by inhibiting the production of cytokines.10,11 37 Anti-inflammatory effects of ginger and it ingredients have also been evaluated in individual 38 organs, such as liver, brain, lung, and colon.12 For example, 6-shogaol suppresses the 3 39 microglial activation in an in vivo neuroinflammatory model and shows a neuroprotective 40 effect in transient global ischemia.13,14 Zingerone, a phenolic alkanone of ginger extract, 41 attenuates LPS-induced acute lung injury and hepatic injury in mice.15 Moreover, zingerone 42 improves experimental colitis in mice via nuclear factor-κB (NF-κB) activity in our previous 43 study.16 However, these studies raise a question: do ginger extract and its constituents exhibit 44 broad-spectrum anti-inflammatory effects in most organs? 45 To address this question, we applied bioluminescent imaging on LPS-induced transgenic 46 mice, which carried NF-κB-driven luciferase genes, to comprehensively monitor the 47 anti-inflammatory effects of ginger and zingerone in whole body and organs. NF-κB 48 bioluminescent imaging has been applied to assess host responses to the implantation of 49 biomaterials and the exposure of ionizing radiation.17,18 It has been used to evaluate the 50 anti-inflammatory potentials of vanillin and ginger extract on experimental colitis.16 It also 51 has been utilized to monitor the anti-inflammatory effects of medicinal herbs on LPS-induced 52 acute systemic inflammation and carbon tetrachloride-induced chronic hepatitis.19,20 53 Immunohistochemical 54 anti-inflammatory mechanisms of ginger and zingerone. Our findings suggested that ginger 55 and zingerone were likely to be broad-spectrum anti-inflammatory agents in most organs that 56 suppressed the activation of NF-κB, the production of interleukin-1β (IL-1β), and the 57 infiltration of inflammatory cells. (IHC) staining was 4 further performed to elucidate the 58 59 60 MATERIALS AND METHODS Chemicals. LPS (from Escherichia coli 055:B5), zingerone, and 61 3-(4,5-dimethylthiazol-2-yl-2,5-diohenyl tetrazolium bromide (MTT) were purchased from 62 Sigma (St. Louis, MO). MG-132, a NF-κB inhibitor, was purchased from Santa Cruz (Dallas, 63 TX). D-Luciferin was purchased from Xenogen (Hopkinton, MA). Mouse monoclonal 64 antibody against p65 was purchased from Chemicon (Temecula, CA). Rabbit polyclonal 65 antibodies against IL-1β and CD11b were purchased from Santa Cruz (Dallas, TX) and 66 Abcam (Cambridge, UK), respectively. 67 68 Preparation of Ginger Extract. Dried ginger was purchased from Xin Lung Chinese 69 Herbal Medicine Pharmacy (Taichung, Taiwan). The voucher specimen has been deposited in 70 Graduate Institute of Chinese Medicine, China Medical University. Ginger was ground to a 71 fine powder and extracted by mixing 20 g powder with 100 ml ethanol at room temperature 72 with shaking. Three days later, the supernatant was collected and stored at -30C for further 73 analysis. 74 75 Cell Culture. Recombinant HepG2/NF-κB cells, which carried NF-κB-driven luciferase 76 genes, were constructed previously.19 HepG2/NF-κB cells were maintained in Dulbecco's 5 77 modified Eagle's medium (Life Technologies, Gaithersburg, MD) supplemented with 10% 78 fetal bovine serum (Hyclone, Logan, Utah) and incubated at 37C with 5% CO2. 79 80 Luciferase Assay and Cell Viability Assay. HepG2/NF-κB cells (2×107 cells) were 81 seeded in a 96-well plate and incubated at 37C overnight. LPS (100 ng/ml), MG-132 (5 μM), 82 or various amounts of ginger and zingerone were then added to cells and incubated at 37C 83 for 24 h. Cell viability was analyzed by MTT colorimetric assay as described previously.19 84 Luciferase assay was performed as described previously.19 Relative NF-κB activity was 85 calculated by dividing the relative luciferase unit (RLU) of compound-treated cells by the 86 RLU of solvent-treated cells. 87 88 Animal Experiments. Transgenic mice, carrying the luciferase genes driven by 89 NF-B-responsive elements, were constructed as described previously.17 Mouse experiments 90 were conducted under ethics approval from China Medical University Animal Care and Use 91 Committee (Permit No. 97-28-N). 92 Six-week-old female transgenic mice were randomly divided into four groups of five mice: 93 (1) mock, no treatment; (2) LPS, (3) LPS/ginger, and (4) LPS/zingerone. Mice were 94 challenged intraperitoneally with 1 mg/kg LPS and then orally with 100 mg/kg ginger extract 95 or zingerone 10 min later. Four hours later, mice were imaged for the luciferase activity and 6 96 subsequently sacrificed for ex vivo imaging and IHC staining. 97 98 In Vivo and Ex Vivo Bioluminescence Imaging. Bioluminescence imaging was 99 performed as described previously.17 Briefly, for in vivo imaging, mice were injected 100 intraperitoneally with 150 mg/kg D-luciferin, placed in the IVIS Imaging System® 200 Series 101 chamber (Xenogen, Hopkinton, MA) 5 min later, and imaged for 1 min. Photons emitted 102 from bodies were quantified using Living Image® software (Xenogen, Hopkinton, MA).The 103 intensity of the signal from bodies was quantified as the sum of all photon counts per second 104 and presented as photon/sec. For ex vivo imaging, mice were injected with D-luciferin and 105 sacrificed 5 min later. The organs were removed immediately, placed in the IVIS chamber, 106 and imaged for 1 min. The intensity of signal was quantified as the sum of all detected photon 107 counts per second with the region of interest and presented as photon/sec/cm2/steradian (sr). 108 109 Cytokine Enzyme-Linked Immunosorbent Assay (ELISA). The amounts of 110 proinflammatory cytokines, including IL-1β and tumor necrosis factor-α (TNF-α), in sera 111 were quantified using Quantikine® Mouse ELISA kits (R&D Systems, Minneapolis, MN). 112 Briefly, mouse sera were added to wells coated with anti-IL-1β or anti-TNF-α antibodies and 113 incubated at room temperature for 2 h. Biotinylated anti-mouse IL-1β or TNF-α antibodies, 114 and avidin-horseradish peroxidase were added sequentially to wells. After a final wash, 7 115 chromogenic substrate (tetramethylbenzidine) was added and the reaction was stopped with 2 116 N H2SO4. The absorbance at 450 nm was measured using an ELISA reader (Multiskan GO, 117 Thermo Scientific, Waltham, MA). 118 119 IHC Staining. Parafilm-embedded small intestines were cut into 5-µm-thick sections, 120 deparaffinized in xylene, and rehydrated in graded ethanol. Sections were incubated with 121 anti-p65, anti-IL-1β, or anti-CD11b antibodies overnight at 4C and then incubated with a 122 biotinylated secondary antibody (Zymed Laboratories, Carlsbad, CA) for 20 min at room 123 temperature. Finally, the sections were incubated with avidin-biotin complex reagent and 124 stained with 3,3'-diaminobenzidine according to manufacturer’s protocol (Histostain®-Plus, 125 Zymed Laboratories, Carlsbad, CA). 126 127 Statistics Analysis. Data were presented as mean ± standard error. Student’s t-test was 128 used for the comparison between two experiments. A value of p < 0.05 was considered 129 statistically significant. 130 131 132 133 RESULTS Ginger and Zingerone Suppressed LPS-Induced NF-κB Activities in Cells. Zingerone 8 134 is a phenolic alkanone of ginger extract (Figure 1), and the content of zingerone was 135 approximately 0.01 mg/ml in ethanolic extract of ginger by high-performance liquid 136 chromatography analysis (see Supplementary Figure 1 of the Supporting Information). We 137 first analyzed the effects of ginger and zingerone on LPS-induced NF-κB activation in cells. 138 HepG2/NF-κB cells were treated with LPS, followed by MG-132 or various amounts of 139 ginger and zingerone. As shown in Figure 2, LPS increased the NF-κB activity by 4-fold, 140 compared with mock. MG-132, a well-known NF-κB inhibitor, significantly suppressed 141 LPS-induced NF-κB activities. Ginger and zingerone decreased NF-κB activities induced by 142 LPS, and the decrease displayed a dose-dependent manner. The maximal inhibition 143 (84.5±3.5% and 96.2±0.6%) was observed at 100 μg/ml ginger and zingerone, respectively. 144 Moreover, zingerone was more effective than ginger on the inhibition of LPS-induced NF-κB 145 activity. No visible cytotoxic effects were observed, judged by MTT assay (data not shown). 146 These findings suggested that ginger and zingerone significantly suppressed NF-κB activities 147 induced by LPS in cells. 148 149 Ginger and Zingerone Suppressed LPS-Induced Inflammation in Mice. The in vivo 150 anti-inflammatory effects of ginger and zingerone were then analyzed by NF-κB 151 bioluminescent imaging. Ginger extract or zingerone was orally given to transgenic mice, 152 which has been challenged by 1 mg/kg LPS. The NF-κB-dependent bioluminescence was 9 153 monitored 4 h later. As shown in Figure 3, LPS induced an approximately 4-fold increase in 154 NF-κB-driven luminescent intensity, compared with mock. The induced luminescence was 155 observed over the whole body, and the strongest luminescence appeared in the abdominal 156 region. Ginger and zingerone significantly decreased the LPS-induced luminescent intensity 157 by 26.9±14.3% and 38.5±6.2%, respectively. These findings suggested that ginger and 158 zingerone suppressed LPS-induced NF-κB-dependent luminescence in mice. 159 NF-κB plays a crucial role in the regulation of immunity. We wondered whether the 160 intensity of NF-κB-driven luminescence was correlated with inflammation. The amount of 161 proinflammatory cytokines, including IL-1β and TNF-α, in sera were therefore quantified by 162 ELISA. As shown in Figure 4, LPS significantly increased the amount of IL-1β and TNF-α 163 in sera by 55.6±7.6 and 172±57.5 fold, respectively. However, ginger and zingerone 164 significantly decreased LPS-induced IL-1β and TNF-α production in sera. Ginger reduced the 165 production of IL-1β and TNF-α by 73.5±23% and 63.9±18.2%, respectively, while zingerone 166 decreased IL-1β and TNF-α production by 79.9±10.4% and 81.3±6.2%, respectively. These 167 data suggested that ginger and zingerone suppressed LPS-induced systemic inflammation in 168 mice. Moreover, the correlation between NF-κB-dependent luminescent intensity and 169 cytokine production indicated the representative of NF-κB-driven luminescence on the degree 170 of inflammation. 171 10 172 Ginger and Zingerone Suppressed LPS-Induced Inflammation in Most Organs. 173 Previous studies have shown that ginger or zingerone displayed anti-inflammatory efficacies 174 in individual organs, such as liver, lung, brain, and colons. We would like to know whether 175 ginger and zingerone exhibited broad-spectrum anti-inflammatory actions in most organs. 176 Transgenic mice were therefore challenged with LPS and orally administered with ginger 177 extract and zingerone, and NF-κB-driven luminescence in organs was monitored 4 h later. As 178 shown in Figure 5, luminescent intensities of organs were increased by LPS, suggesting that 179 intraperitoneal injection of LPS induced inflammation in most organs. The maximal 180 induction of luminescence by LPS was observed in kidney (15.9±8 fold), followed by 181 intestine (15.2±6 fold), brain (10.4±1.4 fold), heart (5.7±1.5 fold), liver (4.7±1.9 fold), spleen 182 (4.7±1.2 fold), lung (3.7±1.4 fold), and stomach (2.7±0.7 fold). Administration of ginger and 183 zingerone significantly decreased LPS-induced NF-κB-driven luminescence in most organs. 184 The maximal inhibition of LPS-induced luminescence by ginger and zingerone was observed 185 in intestine, followed by kidney, liver, heart, and brain. Zingerone significantly decreased 186 LPS-induced luminescence in lung, while ginger slightly decreased luminescent signals in 187 lung. Moreover, both ginger and zingerone slightly decreased LPS-induced luminescent 188 intensities in spleen. 189 We further analyzed the anti-inflammatory effects of ginger and zingerone in different 190 segments of small intestine. We divided the small intestine into 35 segments and the length 11 191 ratio of duodenum, jejunum, and ileum was 1:3:2. The intensity of signal from each segment 192 was quantified as photon/sec. The suppression of LPS-induced luminescent signal by ginger 193 or zingerone was further represented as the inhibitory percentage. As shown in Figure 6, LPS 194 increased NF-κB-driven luminescence in whole small intestine, and the strong luminescence 195 was observed from segment 17 to 30, which corresponded to the region between mid-jejunum 196 and mid-ileum. Both ginger and zingerone suppressed LPS-induced bioluminescent intensity 197 in whole small intestine, and the inhibition from segment 14 to 35 was > 50% by ginger and 198 zingerone. Overall, these data suggested that ginger and zingerone displayed broad-spectrum 199 anti-inflammatory activities in most organs. Additionally, ex vivo imaging first showed that 200 LPS induced a more severe inflammation in the region spanning from mid-jejunum to 201 mid-ileum. Moreover, LPS-induced luminescent signal in the junction of jejunum and ileum 202 was suppressed efficiently by ginger and zingerone. 203 204 Ginger and Zingerone Inhibited LPS-Induced NF-κB Activation, IL-1β Production, 205 and Inflammatory Cell Infiltration in Small Intestine. IHC staining was further performed 206 to analyze the anti-inflammatory effects and mechanisms of ginger and zingerone in small 207 intestine. As shown in Figure 7, the number of IL-1β-positive cells was increased by LPS, 208 compared with mock. However, the expression of IL-1β-positive area was decreased by 209 ginger and zingerone. In addition, LPS increased the number of CD11b-positive cells, 12 210 including monocytes and granulocytes, while ginger and zingerone inhibited the expression 211 of CD11b-positive area. These data suggested that ginger and zingerone suppressed the 212 production of IL-1β and the infiltration of inflammatory cells, resulting in the amelioration of 213 LPS-inflammation in small intestine. 214 Because NF-κB plays a critical role in inflammation, we analyzed the level of NF-κB 215 activity by IHC staining. The monoclonal antibody used here was against p65 nuclear 216 localization sequence, which was blocked by inhibitory IκB when NF-κB was inactivated. 217 LPS increased the number of p65-positive cells, while ginger and zingerone decreased the 218 p65-positive area. These findings suggested the inhibition of ginger and zingerone on 219 LPS-induced inflammation might be through NF-κB signaling pathway. 220 221 222 DISCUSSION 223 In this study, we comprehensively evaluated the anti-inflammatory effects of ginger and 224 zingerone by NF-κB bioluminescent imaging. Because of the light absorption by pigmented 225 molecules and the low spatial resolution of bioluminescent imaging, we performed ex vivo 226 imaging to monitor the effects of ginger and zingerone on individual organs. Previous studies 227 have shown that ginger extract and zingerone display anti-inflammatory activities in specific 228 organs. For example, ginger extracts ameliorate LPS-induced hepatic injury and experimental 13 229 colitis in mice.16,22 Administration of ginger extracts significantly represses paw and joint 230 swelling in rats with severe chronic adjuvant arthritis.23 Ginger also exhibits a protective role 231 on the diabetic brain by modulating the astroglial response to the injury in rats.24 Moreover, 232 zingerone attenuates LPS-induced lung injury in mice.15 By NF-κB bioluminescent imaging, 233 we found that administration of ginger and zingerone inhibited LPS-induced NF-κB-driven 234 luminescence in brain, lung, liver, and colon However, we newly identified that ginger and 235 zingerone rreduced LPS-induced luminescent intensities in heart, stomach, kidney, and small 236 intestine. In addition, the maximal inhibition of LPS-induced luminescent signal by ginger 237 and zingerone was observed in intestines, followed by kidney. The in vivo metabolism or 238 pharmacokinetics of ginger and zingerone has been reported. After oral administration of 2 g 239 ginger extract in human, the glucuronide and sulfate metabolites of ginger components, such 240 as gingerols and shogaol, are detected in plasma and gastrointestinal tract.25 Oral dosage (100 241 mg/kg) of zingerone in rats results in the urinary excretion of glucuronide and/or sulfate 242 conjugates of zingerone.26 The distribution of ginger extract and zingerone in gastrointestinal 243 tract and kidney might explain their anti-inflammatory effects in small intestine and kidney. 244 Because of the correlation between NF-κB-driven luminescent signals and inflammation, we 245 speculated that ginger and zingerone exhibited broad-spectrum anti-inflammatory activities 246 that suppressed the LPS-induced inflammation in various organs. 247 Anti-inflammatory mechanisms of ginger and its constituents have been analyzed in in 14 248 vitro and in vivo studies. For instance, ginger extract ameliorates LPS-induced hepatic injury 249 via inhibiting the production of proinflammatory cytokines and attenuating the 250 mitogen-activated protein kinases and NF-κB signaling pathways.22 It also inhibits the 251 activities of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS), and thus 252 inhibits the synthesis of prostaglandins and NO, mediators of inflammation.27 Ginger 253 derivatives, such as 6-shogaol, reduce osteoarthritis symptom by inhibiting toll-like receptor 254 4 (TLR-4)-mediated innate immunity and cathepsin-k activity.28 6-Shogaol also displays a 255 neuroprotective effect via inhibiting iNOS, COX-2, proinflammatory cytokines, and NF-κB 256 activities in LPS-treated microglial cells.14 In addition, 1-dehydro-10-gingerdione inhibits 257 TLR-4-mediated signaling cascades and cytokine expression via blockade of LPS binding to 258 myeloid differentiation protein 2, a co-receptor of TLR-4 in macrophages.29 In this study, we 259 found that ginger and zingerone inhibited LPS-induced NF-κB activities in various organs in 260 mice. Ginger and zingerone also suppressed the nuclear translocation of NF-κB subunit p65, 261 the production of IL-1β, and the infiltration of granulocytes in small intestine. Overall, this 262 study suggested that ginger and zingerone shared a common anti-inflammatory mechanism 263 by inhibiting NF-κB activities and proinflammatory cytokine production in LPS-induced 264 systemic inflammation. Moreover, ginger exhibits a non-steroid anti-inflammatory activity 265 via inhibiting the activities of COX-2 and iNOS in other studies, probably explaining why 266 ginger displayed a broad-spectrum anti-inflammatory activity in various organs in this study. 15 267 Detailed anti-inflammatory effects of ginger and zingerone in small intestine were 268 evaluated here. It is interesting to find that LPS increased NF-κB-driven luminescence in the 269 entire small intestine, especially from the middle portion of jejunum to the end of ileum. It is 270 known that LPS activates macrophages, neutrophils, and dendritic cells via binding to TLR-4 271 and activating downstream NF-κB activity. The activated immune cells then initiate the 272 inflammatory response and present antigens to lymphocytes in lymph nodes.30 In comparison 273 with duodenum, jejunum and ileum have abundant Peyer's patches, organized lymphoid 274 nodules, in mice. Thus, we speculated that LPS induced maximal NF-κB-driven 275 luminescence in jejunum and ileum might result from the abundance of Peyer's patches. We 276 also found that ginger and zingerone suppressed LPS-induced NF-κB-dependent 277 bioluminescent signals in the entire small intestine, especially in the region between 278 mid-jejunum and mid-ileum. Zingerone possesses the vanillyl moiety, which is considered 279 important for the activation of vanilloid receptor 1 (VR1) expressed in nociceptive sensory 280 neurons.31 Recent study shows that activated VR1 protects against LPS-mediated renal injury 281 possibly via reducing renal inflammation responses.32 In addition, sensory VR1 has been 282 found to modulate cytokine response to LPS and thereby induce the subsequent 283 anti-inflammatory effect in the gut mucosa.33 VR1 nerve fibers are observed within enteric 284 ganglia of jejunum and ileum,34 probably explaining why zingerone displayed more activities 285 in jejunum and ileum than in duodenum. 16 286 In conclusion, we comprehensively evaluated the anti-inflammatory effects of ginger and 287 zingerone on LPS-induced systemic inflammation via NF-κB bioluminescent imaging. Our 288 data showed for the first time that ginger and zingerone suppressed NF-κB-drive 289 luminescence in most organs. In addition, our findings suggested that ginger and zingerone 290 were likely to be broad-spectrum anti-inflammatory agents in most organs that suppressed the 291 activation of NF-κB, the production of IL-1β, and the infiltration of inflammatory cells. 292 293 294 ABBREVIATIONS USED 295 COX-2, 296 immunohistochemical; iNOS, inducible nitric oxide synthase; IL-1β, interleukin-1β; LPS, 297 lipopolysaccharide; MTT, 3-(4,5-dimethylthiazol-2-yl-2,5-diohenyl tetrazolium bromide; NO, 298 nitric oxide; NF-κB, nuclear factor-κB; RLU, relative luciferase unit; sr, steradian; TLR-4, 299 toll-like receptor 4; TNF-α, tumor necrosis factor-α; VR1, vanilloid receptor 1 cyclooxygenase-2; ELISA, enzyme-linked 300 301 SUPPORTING INFORMATION 302 HPLC profile of ginger extract (Supplementary Figure S1). 303 304 FUNDING SOURCES 17 immunosorbent assay; IHC, 305 This work was supported by grants from Ministry of Science and Technology 306 (NSC101-2320-B-039-034-MY3, NSC102-2632-B-039-001-MY3, 307 104-2815-C-039-012-B), Medical 308 CMU103-SR-44), and CMU under the Aim for Top University Plan of the Ministry of 309 Education, Taiwan. China 310 311 NOTES 312 The authors declare no competing financial interest. 313 18 University and (CMU102-NSC-04 MOST and 314 315 REFERENCES (1) Ryan, J. L.; Heckler, C. E.; Roscoe, J. A.; Dakhil, S. R.; Kirshner, J.; Flynn, P. J.; 316 Hickok, J. T.; Morrow, G. R. Ginger (Zingiber officinale) reduces acute 317 chemotherapy-induced nausea: a URCC CCOP study of 576 patients. Support. Care Cancer 318 2012, 20, 1479-1489. 319 (2) Dabaghzadeh, F.; Khalili, H.; Dashti-Khavidaki, S.; Abbasian, L.; Moeinifard, A. 320 Ginger for prevention of antiretroviral-induced nausea and vomiting: a randomized clinical 321 trial. Expert Opin. Drug Saf. 2014, 13, 859-866. 322 (3) Chopra, A.; Saluja, M.; Tillu, G.; Sarmukkaddam, S.; Venugopalan, A.; Narsimulu, G.; 323 Handa, R.; Sumantran, V.; Raut, A.; Bichile, L.; Joshi, K.; Patwardhan, B. Ayurvedic 324 medicine offers a good alternative to glucosamine and celecoxib in the treatment of 325 symptomatic knee osteoarthritis: a randomized, double-blind, controlled equivalence drug 326 trial. Rheumatology 2013, 52, 1408-1417. 327 (4) Bartels, E. M.; Folmer, V. N.; Bliddal, H.; Altman, R. D.; Juhl, C.; Tarp, S.; Zhang, W.; 328 Christensen, R. Efficacy and safety of ginger in osteoarthritis patients: a meta-analysis of 329 randomized placebo-controlled trials. Osteoarthr. Cartil. 2015, 23, 13-21. 330 331 (5) Black, C. D.; Herring, M. P.; Hurley, D. J.; O'Connor, P. J. Ginger (Zingiber officinale) reduces muscle pain caused by eccentric exercise. J. Pain 2010, 11, 894-903. 19 332 (6) Rahnama, P.; Montazeri, A.; Huseini, H. F.; Kianbakht, S.; Naseri, M. Effect of 333 Zingiber officinale R. rhizomes (ginger) on pain relief in primary dysmenorrhea: a placebo 334 randomized trial. BMC Complement. Altern. Med. 2012, 12, 92. 335 (7) Mozaffari-Khosravi, H.; Talaei, B.; Jalali, B. A.; Najarzadeh, A.; Mozayan, M. R. The 336 effect of ginger powder supplementation on insulin resistance and glycemic indices in 337 patients with type 2 diabetes: a randomized, double-blind, placebo-controlled trial. 338 Complement. Ther. Med. 2014, 22, 9-16. 339 (8) Jung, H. W.; Yoon. C. H.; Park, K. M.; Han, H. S.; Park, Y. K. Hexane fraction of 340 Zingiberis Rhizoma Crudus extract inhibits the production of nitric oxide and 341 proinflammatory cytokines in LPS-stimulated BV2 microglial cells via the NF-kappaB 342 pathway. Food Chem. Toxicol. 2009, 47, 1190-1197. 343 (9) Podlogar, J. A.; Verspohl, E. J. Antiinflammatory effects of ginger and some of its 344 components in human bronchial epithelial (BEAS-2B) cells. Phytother. Res. 2012, 26, 345 333-336. 346 (10) Li, F.; Nitteranon, V.; Tang, X.; Liang, J.; Zhang, G.; Parkin, K. L.; Hu, Q. In vitro 347 antioxidant and anti-inflammatory activities of 1-dehydro-[6]-gingerdione, 6-shogaol, 348 6-dehydroshogaol and hexahydrocurcumin. Food Chem. 2012, 135, 332-327. 20 349 (11) Huang, S. H.; Lee, C. H.; Wang, H. M.; Chang, Y. W.; Lin, C. Y.; Chen, C. Y.; Chen, 350 Y. H. 6-Dehydrogingerdione restrains lipopolysaccharide-induced inflammatory responses in 351 RAW 264.7 macrophages. J. Agric. Food Chem. 2014, 62, 9171-9179. 352 (12) Rahmani, A. H.; Shabrmi, F. M.; Aly, S. M. Active ingredients of ginger as potential 353 candidates in the prevention and treatment of diseases via modulation of biological activities. 354 Int. J. Physiol. Pathophysiol. Pharmacol. 2014, 6, 125-136. 355 (13) Ha, S. K.; Moon, E.; Ju, M. S.; Kim, D. H.; Ryu, J. H.; Oh, M. S.; Kim, S. Y. 356 6-Shogaol, a ginger product, modulates neuroinflammation: a new approach to 357 neuroprotection. Neuropharmacology 2012, 63, 211-223. 358 (14) Moon, M.; Kim, H. G.; Choi, J. G.; Oh, H.; Lee, P. K.; Ha, S. K.; Kim, S. Y.; Park, 359 Y.; Huh, Y.; Oh, M. S. 6-Shogaol, an active constituent of ginger, attenuates 360 neuroinflammation and cognitive deficits in animal models of dementia. Biochem. Biophys. 361 Res. Commun. 2014, 449, 8-13. 362 (15) Xie, X.; Sun, S.; Zhong, W.; Soromou, L. W.; Zhou, X.; Wei, M.; Ren, Y.; Ding, Y. 363 Zingerone attenuates lipopolysaccharide-induced acute lung injury in mice. Int. 364 Immunopharmacol. 2014, 19, 103-109. 365 (16) Hsiang, C. Y.; Lo, H. Y.; Huang, H. C.; Li, C. C.; Wu, S. L.; Ho, T. Y. Ginger extract 366 and zingerone ameliorated trinitrobenzene sulphonic acid-induced colitis in mice via 367 modulation of nuclear factor-κB activity and interleukin-1β signalling pathway. Food Chem. 21 368 2013, 136, 170-177. 369 (17) Ho, T. Y.; Chen, Y. S.; Hsiang, C. Y. Noninvasive nuclear factor-κB 370 bioluminescence imaging for the assessment of host-biomaterial interaction in transgenic 371 mice. Biomaterials 2007, 28, 4370-4377. 372 (18) Chang, C. T.; Lin, H.; Ho, T. Y.; Li, C. C.; Lo, H. Y.; Wu, S. L.; Huang, Y. F.; Liang, 373 J. A.; Hsiang, C. Y. Comprehensive assessment of host responses to ionizing radiation by 374 nuclear factor-κB bioluminescence imaging-guided transcriptomic analysis. PLoS ONE 2011, 375 6, e23682. 376 (19) Li, C. C.; Hsiang, C. Y.; Lo, H. Y.; Pai, F. T.; Wu, S. L.; Ho, T. Y. Genipin inhibits 377 lipopolysaccharide-induced acute systemic inflammation in mice as evidenced by nuclear 378 factor-κB bioluminescent imaging-guided transcriptomic analysis. Food Chem. Toxicol. 2012, 379 50, 2978-2986. 380 (20) Li, C. C.; Hsiang, C. Y.; Wu, S. L.; Ho, T. Y. 2012. Identification of novel 381 mechanisms of silymarin on the carbon tetrachloride-induced liver fibrosis in mice by nuclear 382 factor-κB bioluminescent imaging-guided transcriptomic analysis. Food Chem. Toxicol. 2012, 383 50, 1568-1575. 384 385 (21) Badr, C. E. Bioluminescence imaging: basics and practical limitations. Methods Mol. Biol. 2014, 1098, 1-18. 22 386 (22) Choi, Y. Y.; Kim, M. H.; Hong, J.; Kim, S. H.; Yang, W. M. Dried ginger (Zingiber 387 officinalis) inhibits inflammation in a lipopolysaccharide-induced mouse model. Evid. Based 388 Complement. Alternat. Med. 2013, 2013, 914563. 389 390 391 392 393 394 395 396 397 398 (23) Sharma, J. N.; Srivastava, K. C.; Gan, E. K. Suppressive effects of eugenol and ginger oil on arthritic rats. Pharmacology 1994, 49, 314-318. (24) El-Akabawy, G.; El-Kholy, W. Neuroprotective effect of ginger in the brain of streptozotocin-induced diabetic rats. Ann. Anat. 2014, 196, 119-128. (25) Yu, Y.; Zick, S.; Li, X.; Zou, P.; Wright, B.; Sun, D. Examination of the pharmacokinetics of active ingredients of ginger in humans. AAPS J. 2011, 13, 417-426. (26) Monge, P.; Scheline, R.; Solheim, E. The metabolism of zingerone, a pungent principle of ginger. Xenobiotica 1976, 6, 411-423. (27) Yu, Y. S.; Hsu, C. L.; Yen, G. C. Anti-inflammatory effects of the roots of Alpinia pricei Hayata and its phenolic compounds. J. Agric. Food Chem. 2009, 57: 7673-7680. 399 (28) Villalvilla, A.; da Silva, J. A.; Largo, R.; Gualillo, O.; Vieira, P. C.; 400 Herrero-Beaumont, G.; Gómez, R. 6-Shogaol inhibits chondrocytes' innate immune responses 401 and cathepsin-K activity. Mol. Nutr. Food Res. 2014, 58, 256-266. 402 (29) Park, S. H.; Kyeong, M. S.; Hwang, Y.; Ryu, S. Y.; Han, S. B.; Kim, Y. Inhibition of 403 LPS binding to MD-2 co-receptor for suppressing TLR4-mediated expression of 23 404 inflammatory cytokine by 1-dehydro-10-gingerdione from dietary ginger. Biochem. Biophys. 405 Res. Commun. 2012, 419, 735-740. 406 407 (30) Morris, M. C.; Gilliam, E. A.; Li, L. Innate immune programing by endotoxin and its pathological consequences. Front Immunol. 2015, 5, 680. 408 (31) Dedov, V. N.; Tran, V. H.; Duke, C. C.; Connor, M.; Christie, M. J.; Mandadi, S.; 409 Roufogalis, B. D. Gingerols: a novel class of vanilloid receptor (VR1) agonists. Br. J. 410 Pharmacol. 2002, 137, 793-798. 411 (32) Wang, Y.; Wang, D. H. Deletion of the transient receptor potential vanilloid type 1 412 channel exacerbates renal inflammation induced by lipopolysaccharide in mice. FASEB J. 413 2013, 27, 721.10 414 (33) Assas, B. M.; Miyan, J. A.; Pennock, J. L. Cross-talk between neural and immune 415 receptors provides a potential mechanism of homeostatic regulation in the gut mucosa. 416 Mucosal Immunol. 2014, 7, 1283-1289. 417 (34) Vinuesa, A. G.; Sancho, R.; García-Limones, C.; Behrens, A.; ten Dijke, P.; Calzado, 418 M. A.; Muñoz, E. Vanilloid receptor-1 regulates neurogenic inflammation in colon and 419 protects mice from colon cancer. Cancer Res. 2012, 72, 1705-1716. 24 420 FIGURE CAPTIONS 421 Figure 1. Ginger and zingerone. (A) Morphology of whole parts and cross sections of dried 422 ginger. (B) Chemical structure of zingerone. 423 424 Figure 2. Effects of ginger and zingerone on LPS-induced NF-κB activities in cells. 425 HepG2/NF-κB cells were treated with 100 ng/ml LPS and/or various amounts of ginger 426 extract and zingerone. MG-132 (5 µM) was used as a positive control. Twenty-four hours 427 later, NF-κB activity was measured by luciferase assay. Results are expressed as relative 428 NF-κB activity, which is presented as the comparison with RLU relative to solvent-treated 429 cells. Values are mean ± standard error (n=6). ###p < 0.001, compared with mock. **p < 430 0.01, ***p < 0.001, compared with LPS. 431 432 Figure 3. NF-κB-driven luminescence in living mice. (A) In vivo image. Transgenic mice 433 were administered with 1 mg/kg LPS and then treated with 100 mg/kg ginger or zingerone. 434 Four hours later, mice were injected intraperitoneally with D-luciferin and imaged for 1 min. 435 The color overlay on the image represents the photon/sec emitted from mice, as indicated by 436 the color scale. Photos are representative images (n=5/group). (B) Quantification of photon 437 emission from the whole body. Values are mean ± standard error. ###p < 0.001, compared 438 with mock. *p < 0.05, ***p < 0.001, compared with LPS. 25 439 440 Figure 4. Effects of ginger and zingerone on LPS-induced IL-1β and TNF-α production in 441 sera. Transgenic mice were challenged with 1 mg/kg LPS and then given with 100 mg/kg 442 ginger or zingerone. Four hours later, mice were sacrificed and the amount of IL-1β (A) and 443 TNF-α (B) in sera was quantified by ELISA. Values are mean ± standard error (n=5/group). 444 ##p < 0.01, compared with mock. *p < 0.05, **p < 0.01, ***p < 0.001, compared with LPS. 445 446 Figure 5. NF-κB-dependent luminescence in individual organs. (A) Ex vivo imaging. 447 Transgenic mice were administered with 1 mg/kg LPS and then treated with 100 mg/kg 448 ginger or zingerone. Four hours later, mice were injected intraperitoneally with D-luciferin. 449 Five minutes later, mice were sacrificed, and organs were excised rapidly and subjected to 450 image (n=5/group). (B) Quantification of photon emission from organs. Values are mean ± 451 standard error. ##p < 0.01, ###p < 0.001, compared with mock. *p < 0.05, **p < 0.01, ***p < 452 0.001, compared with LPS. 453 454 Figure 6. Anti-inflammatory effects of ginger and zingerone in various segments of small 455 intestine. Transgenic mice were challenged with 1 mg/kg LPS and then given with 100 mg/kg 456 ginger or zingerone. Four hours later, mice were injected intraperitoneally with D-luciferin. 457 Five minutes later, mice were sacrificed, and organs were excised rapidly and subjected to 26 458 image. Small intestine was divided equally into 35 segments and the photon emission from 459 each segment was quantitated. Results are expressed as inhibition (%). Heatmap of small 460 intestine is shown on the bottom. The color overlay on the heatmap represents the photon/sec 461 emitted from each segment of small intestine, as indicated by the color scale. 462 463 Figure 7. IHC staining analysis of p65, IL-1β, and CD11b in jejunum. Transgenic mice were 464 challenged with 1 mg/kg LPS and then given with 100 mg/kg ginger or zingerone. Four hours 465 later, mice were sacrificed. Section of mid-jejunum was stained with antibodies against p65, 466 IL-1β, and CD11b (400× magnification). Photos are representative images (n=5/group) 27 Figure 1 (A) (B) 28 Figure 2 5 Ginger Relative NF-κB activity ### Zingerone ** 4 *** *** *** 3 *** *** *** *** 2 *** 1 *** *** *** *** 0 Mock LPS MG-132 0.5 1 2.5 5 10 Concentration (μg/ml) 29 50 100 Figure 3 (A) Mock LPS LPS/Ginger LPS/Zingerone photon/sec (B) ### 400 Total flux (×106 photon/sec) 350 * 300 250 *** 200 150 100 50 0 Mock LPS LPS/Ginger 30 LPS/Zingerone Figure 4 (A) 800 ## IL-1β concetration (pg/ml) 700 600 500 400 300 ** *** 200 100 0 Mock LPS LPS/Ginger LPS/Zingerone (B) 1,800 ## TNF-α concetration (pg/ml) 1,600 1,400 1,200 1,000 800 * 600 ** 400 200 0 Mock LPS LPS/Ginger 31 LPS/Zingerone Figure 5 (A) Brain Heart Lung Liver Spleen Stomach Kidney Intestine Mock LPS LPS/Ginger LPS/Zingerone (B) 25000 ### Mock LPS 20000 LPS/Ginger Luminescent intensity (×103 photon/sec/cm2/sr) LPS/Zingerone 15000 * ### ### 10000 5000 ** ### * *** * ** ** * ### ## * ### ### ** 0 Brain Heart Lung Liver 32 Spleen Stomach Kidney Intestine 0 33 ROI 35 ROI 34 ROI 33 ROI 32 ROI 31 ROI 30 ROI 29 ROI 28 ROI 27 ROI 26 ROI 25 ROI 24 ROI 23 ROI 22 ROI 21 ROI 20 ROI 19 ROI 18 ROI 17 ROI 16 ROI 15 ROI 14 ROI 13 ROI 12 ROI 11 ROI 10 ROI 9 ROI 8 ROI 7 ROI 6 ROI 5 60 ROI 4 ROI 3 ROI 2 ROI 1 Inhibition (o/o) Figure 6 80 Ginger Zingerone 40 20 Mock LPS LPS/Ginger LPS/Zingerone photon/sec Figure 7 Mock LPS LPS/Ginger p65 IL-1β CD11b 34 LPS/Zingerone