Ginger essential oil prevents nash progression by blocking the nlrp3 inflammasome and remodeling the gut microbiota-lps-tlr4 pathway in mice

Ginger essential oil prevents nash progression by blocking the nlrp3 inflammasome and remodeling the gut microbiota-lps-tlr4 pathway in mice

Play all audios:

Loading...

ABSTRACT BACKGROUND Diet and gut microbiota contribute to non-alcoholic steatohepatitis (NASH) progression. High-fat diets (HFDs) change gut microbiota compositions, induce gut dysbiosis,


and intestinal barrier leakage, which facilitates portal influx of pathogen-associated molecular patterns including lipopolysaccharides (LPS) to the liver and triggers inflammation in NASH.


Current therapeutic drugs for NASH have adverse side effects; however, several foods and herbs that exhibit hepatoprotection could be an alternative method to prevent NASH. METHODS We


investigated ginger essential oil (GEO) against palm oil-containing HFDs in LPS-injected murine NASH model. RESULTS GEO reduced plasma alanine aminotransferase levels and hepatic


pro-inflammatory cytokine levels; and increased antioxidant catalase, glutathione reductase, and glutathione levels to prevent NASH. GEO alleviated hepatic inflammation through mediated NLR


family pyrin domain-containing 3 (NLRP3) inflammasome and LPS/Toll-like receptor four (TLR4) signaling pathways. GEO further increased beneficial bacterial abundance and reduced


NASH-associated bacterial abundance. CONCLUSION This study demonstrated that GEO prevents NASH progression which is probably associated with the alterations of gut microbiota and inhibition


of the LPS/TLR4/NF-κB pathway. Hence, GEO may offer a promising application as a dietary supplement for the prevention of NASH. SIMILAR CONTENT BEING VIEWED BY OTHERS PROBIOTICS SUPPRESS


NONALCOHOLIC STEATOHEPATITIS AND CARCINOGENESIS PROGRESSION IN HEPATOCYTE-SPECIFIC PTEN KNOCKOUT MICE Article Open access 28 September 2022 EVALUATION OF THE ROLE OF KEFIR IN MANAGEMENT OF


NON-ALCOHOLIC STEATOHEPATITIS RAT MODEL VIA MODULATION OF NASH LINKED MRNA-MIRNA PANEL Article Open access 05 January 2023 GUT MICROBIOME DETERMINES THERAPEUTIC EFFECTS OF OCA ON NAFLD BY


MODULATING BILE ACID METABOLISM Article Open access 31 May 2023 INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) stands as the prevailing chronic liver disease on a global scale,


encompassing a broad spectrum of liver conditions [1, 2]. Among these, non-alcoholic steatohepatitis (NASH) represents a distinct entity characterized by hepatic steatosis of at least 5%,


accompanied by inflammation and ballooned hepatocytes, with or without concurrent hepatic fibrosis [3]. If left untreated, NASH can advance to liver cirrhosis and, ultimately, hepatocellular


carcinoma. The concept of the multiple-hit hypothesis recognizes that environmental factors, such as diet, lifestyle, and gut microbiota, can intricately interact with genetic and


epigenetic factors, collectively contributing to the development of liver injury [4]. Typically, NAFLD pathogenesis caused by a high-fat diet (HFD) involves increased hepatic fat


accumulation, oxidative stress, and inflammatory responses [5]. HFD intake results in the elevation of free fatty acid (FFA) levels and stimulates the expression of sterol regulatory


element-binding protein-1c (SREBP1-c)-mediated de novo lipogenesis and 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR)-mediated cholesterol synthesis. Peroxisome proliferator-activated


receptor-α (PPARα), which regulates the oxidation of fatty acids, is expressed at lower levels. These factors promote triglyceride and cholesterol accumulation in the liver [6]. Excessive


hepatic FFAs lead to the upregulation of the expression of the cytochrome P450 isoform 2E1 (CYP2E1) and enhance the production of reactive oxygen species (ROS) [7]. Various liver


antioxidants and enzymes can reduce oxidative stress, such as glutathione (GSH), catalase (CAT), and superoxide dismutase (SOD) [8]. Oxidative stress can also cause chronic hepatic


inflammation. Several studies have confirmed the activation of the NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome (comprising NLRP3, ASC, and pro-caspase-1) in


HFD-induced hepatic steatosis, which induces the production of cytokines interleukin (IL)-1β and -18 and leads to hepatic inflammation and cell death [9, 10]. Diet and gut microbiota


interactions play significant roles in the pathogenesis of NASH [11, 12]. Many recent studies have demonstrated that an unhealthy diet characterized by low fiber, high fat, and high sugar


content can induce gut microbiota shifts resulting in gut microbiota dysbiosis and intestinal barrier leakage, which facilitates the portal influx of pathogen-associated molecular patterns


(PAMPs) (e.g., lipopolysaccharides [LPS] or endotoxins) to the liver [13, 14]. A previous study found that the administration of LPS promoted hepatocarcinogenesis in mice [15]. Gut-derived


bacterial products activate several toll-like receptors (TLRs), including TLR4, in different liver cell types, triggering downstream inflammatory responses and cytokine generation to enhance


NASH progression [16]. LPS can activate the TLR4 signaling pathway, leading to nuclear factor kappa B (NF-κB) intracellular signal transduction and the production of inflammatory cytokines


such as tumor necrosis factor (TNF)-α, IL-6, and IL-1β [17]. Several rodent models for inducing NASH and liver fibrosis have been validated including trans-fat-containing amylin liver NASH


(AMLN) [18,19,20]. Due to the detrimental impact of trans-fats on health, the US Food and Drug Administration (FDA) has implemented regulations prohibiting their use in the food industry


[21,22,23]. Consequently, the trans-fat animal model may not accurately replicate human dietary patterns. The substitution of trans-fat with palm oil in the diet, a palm oil-containing HFD


(P-HFD)–Gubra Amylin NASH (GAN) diet in a mouse model previously confirmed that it leads to an identical phenotype to NASH compared to trans-fat-induced NAFLD mice and demonstrates excellent


clinical translatability [24, 25]. Recently, we developed a NASH C57BL/6 J mouse model using a P-HFD supplemented with intraperitoneal injections of LPS to simulate gut dysbiosis and


endotoxemia. This animal model yielded an equivalent NASH phenotype, intestinal leakage, and endotoxemia, but developed more extra-intestinal microbiota dysbiosis by increasing pathogenic


bacteria and reducing beneficial microbiota [26]. Several approaches have been developed for NASH treatment and prevention, including the use of medications. These NASH therapeutic drugs are


still being developed and have adverse side effects [27]. Several foods and herbs have exhibited hepatoprotective function, and they can be used to prevent NASH [28]. Our previous studies


have demonstrated that a volatile oil from fresh _Zingiber officinale_ Roscoe (ginger), or ginger essential oil (GEO), can potentially alleviate hepatic lipid accumulation, oxidative stress,


and pro-inflammatory cytokines against HFD-induced NAFLD [6]. GEO exerts antibacterial and antifungal effects [29, 30]. Moreover, GEO ameliorates atherosclerosis in ApoE−/− mice by


modulating trimethylamine-N-oxide and gut microbiota [31]. However, the hepatoprotective effects of GEO against NASH and its impact on the gut microbiota, its metabolites, the gut–liver


axis, and NLRP3 inflammasome pathway have not yet been determined. Therefore, this study investigated the hepatoprotective effects of GEO through the NLRP3 inflammasome and gut


microbiota-LPS-TLR4 pathway using a P-HFD with LPS-induced NASH, endotoxemia, and gut dysbiosis in mouse models. METHODS GINGER ESSENTIAL OIL EXTRACTION AND GAS CHROMATOGRAPHY ANALYSIS


Ginger was obtained from the Mingjian Township Farmers’ Association of Nantou County (Taiwan). Raw aged ginger was washed and sliced into small pieces. Distilled water was added to sliced


ginger (1:3) and mixed using a blender. The aged ginger puree was extracted by steam distillation for approximately 6 h. The yellowish GEO was then collected. The GEO was placed in a –20 °C


freezer to exclude water content. The unfrozen liquid was collected and repeatedly placed in the freezer until no frozen liquid portion was detected. The yield of the GEO was 0.14% of the


raw aged ginger. Gas chromatography (GC) was used to analyze the constitution of GEO and its primary compound citral (Sigma-Aldrich Corp., MO, USA). GEO constitution was measured using a


Thermo Scientific Focus GC equipped with an AI 3000 II autosampler, a flame ionization detector, and a Stabilwax (crossbond Carbowax-PEG, Restek) column (60 m × 0.32 mm; 1 μm) [6, 32]. The


GC chromatogram demonstrated that citral is composed of a mixture of geranial and neral geometric isomers. Citral was the main component of GEO, comprising approximately 31% with 18.8% neral


and 12.2% geranial isomers (Supplementary Fig. 1). ANIMAL MODEL Animal experiments were performed according to the guidelines established by the Institutional Animal Care and Use Committee


of National Taiwan University (IACUC) (Approval No: NTU-110-EL-00060). A previous study established a P-HFD with LPS (PL)-induced NASH mouse model in which the severity of gut microbiota


dysbiosis was increased compared to P-HFD alone [26]. Six-week-old male C57BL/6 J mice were purchased from Taiwan National Laboratory Animal Center and housed in an alternate light/dark


cycle (12 h) room at a temperature of 23 ± 2 °C and relative humidity of 50–70%. After 2 weeks of adaptive feeding, the mice were randomly assigned to five groups (_n_ = 7–8/group): ①


control diet (Research Diets, Inc., NJ, USA; D12450K); ② P-HFD (Gubra-Amylin NASH (GAN) diet; Research Diets, Inc., NJ, USA; D09100310) + intraperitoneal injections of LPS (500 µg/kg


bw/week; LPS from Escherichia coli O55:B5; Sigma-Aldrich; L2880) (PL); ③ PL + a low dose of GEO (12.5 mg/kg bw/day) (PL + GL); ④ PL + a medium dose of GEO (62.5 mg/kg bw/day) (PL + GM); and


⑤ PL + a high dose of GEO (125 mg/kg bw/day) (PL + GH). All mice for each treatment were housed in separate cages (_n_ = 3–4 per cage). GEO was administered to the mice through daily oral


gavage. The control diet contained fat (10 kcal%) and carbohydrates, principally corn starch. The P-HFD contained 40 kcal% fat, mainly from palm oil; 20% fructose; and 2% cholesterol [24,


25]. The dosages of GEO selected for this study were based on relevant literature and our previous research, which demonstrated its hepatoprotective effects against NAFLD and alcoholic fatty


liver disease [6, 32]. The dosage of 12.5 mg/kg bw of GEO was considered as the 1X dose in our current investigation. Higher dosages of 62.5 and 125 mg/kg bw were chosen as the 5X and 10X


doses, respectively, to evaluate efficacy across different dosage levels. Body weight and food intake were recorded every week. The mice were sacrificed by carbon dioxide asphyxiation after


12 weeks. Blood was then collected by cardiac punctures using a heparin-coated syringe and transferred to a collection tube. The mice were dissected to extract various specimens. The liver,


spleen, kidney, cecum, the visceral, subcutaneous, epididymal white adipose tissue (WAT) (eWAT), inguinal WAT (iWAT), perirenal WAT (pWAT), mesenteric WAT (mWAT), and inguinal subcutaneous


white adipose tissue (isWAT), and interscapular brown adipose tissue (iBAT) were harvested, imaged, and weighed after sacrificing the mice. The fecal contents of the colons were collected


for DNA sequencing. LIVER HISTOPATHOLOGICAL ANALYSIS The largest right lobe of the liver was fixed in 10% neutral formalin. Histopathological analyses were then performed using the


formalin-fixed paraffin-embedded (FFPE) method. Hepatic histological scoring was performed by the College of Veterinary Medicine Animal Disease Diagnostic Center (National Chung Hsing


University, Taichung, Taiwan). The NAFLD activity score (NAS) was calculated for steatosis, lobular inflammation, and hepatocyte ballooning. Scores of 5–8 were recognized as diagnostic for


NASH [33]. PLASMA BIOCHEMICAL ANALYSES Plasma was extracted by centrifuging the blood at 1000 x _g_ for 15 min at 4 °C. The biochemical parameters of the plasma, including the levels of


total cholesterol, total triglycerides, low-density lipoprotein (LDL), high-density lipoprotein (HDL), aspartate aminotransferase (AST), alanine aminotransferase (ALT), and glucose (TG),


were analyzed using a Beckman Colter AU 5800 (Beckman Colter Inc., USA). LIVER ANTIOXIDANT CAPACITY AND INFLAMMATION ANALYSIS In brief, 0.1 g of liver and 1 mL of homogenization buffer (8 mM


KH2PO4, 12 mM K2HPO4, 1.5% KCl, pH = 7.4) were homogenized, followed by centrifugation at 10,000 x _g_ for 30 min at 4 °C. The supernatant was collected and stored at −80 °C. Liver


homogenates were subjected to assays for GSH, glutathione peroxidase (GPx), glutathione reductase (GRd), superoxide dismutase (SOD), and CAT using commercial kits from Cayman Chemical Co.


(Ann Arbor, MI, USA). Inflammatory cytokine levels in the liver were quantified using enzyme-linked immunosorbent assay (ELISA) kits specific for mouse TNF-α, IL-1β, and IL-6 (Invitrogen,


USA) [26]. WESTERN BLOT ANALYSIS Protein (0.1 g) was extracted with 1 mL of cold lysis buffer (comprising 7 M urea, 2 M thiourea, 2% CHAPS, 0.002% bromophenol blue, 60 mM DTT, and a protease


and phosphatase inhibitor cocktail), followed by centrifugation at 17500 x _g_ for 30 min 4 °C [34]. The supernatants were collected, and the protein concentration was determined using the


Bio-Rad protein assay. Sample buffer (comprising 62.5 mM Tris-HCl, 10% glycerol, 2% SDS, and 0.01% bromophenol blue) was added to the protein samples for protein dilution and boiled at 95 °C


for 10 min. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) was performed to separate the proteins, which were then transferred onto polyvinylidene difluoride (PVDF)


membranes (Millipore Corp., Bedford, MA, USA). The membranes were blocked with 5% bovine serum albumin in Tris-buffered saline with Tween 20 (TBST) buffer (20 mM Tris-base, 150 mM NaCl, and


0.05% Tween 20) for 1 h and washed three times with TBST buffer for 5 min each. The membranes were incubated overnight at 4 °C with primary antibodies against SREBP-1c, HMGCR, PPARα, CYP2E1,


GAPDH (GeneTex, San Antonio, TX, USA), NLRP3, ASC (Abcam, Cambridge, England), Caspase-1 (Santa Cruz Biotechnology, Santa Cruz, CA, USA), TLR4, and NF-κB (Cell Signaling Technology,


Beverly, MA, USA). After removing the unbound primary antibodies with TBST buffer, the membranes were incubated with a horseradish peroxidase-conjugated secondary antibody (GeneTex) at room


temperature for 2 h, then washed. An enhanced chemiluminescence (ECL) substrate (PerkinElmer Life Sciences, Boston, MA, USA) was added to the membranes to detect the enzyme-conjugated


antibodies. The signals were captured using a UVP BioSpectrum AC imaging system (UVP, Upland, CA, USA). Protein expression was quantified by densitometry using the ImageJ software (version


1.47; NIH, Bethesda, MD, USA). The density of each protein band was normalized to that of the control GAPDH protein. FECAL DNA EXTRACTION, 16 S RRNA AMPLICON SEQUENCING, AND SEQUENCING


ANALYSES Mouse fecal samples were removed from the colons during the sacrifices, snap-frozen using liquid N2, and stored at −80 °C before use. Genomic DNA from the feces was extracted using


the QIAmp Power Fecal Pro DNA Kit (QIAGEN, Netherlands) and quantified using a NanoDrop ND-1000 spectrophotometer (Thermo Fisher Scientific). The V3–V4 hypervariable region of the 16 S rRNA


gene was amplified using the following primer pair: forward: 5′-TCG TCG GCA GCG TCA GAT GTG TAT AAG AGA CAG CCT ACG GGN GGC WGC AG-3′ and reverse: 5′-GTC TCG TGG GCT CGG AGA TGT GTA TAA GAG


ACA GGA CTA CHV GGG TAT CTA ATC C-3′. PCR amplification was carried out in a 25-μL reaction mixture containing 5 ng of DNA template, 0.2 μM of forward and reverse primers, and 12.5 μL of 2×


Taq Master Mix (KAPA HiFi HotStart ReadyMix, Roche, Switzerland). The PCR conditions included a cycle at 95 °C for 3 min, followed by 25 cycles at 95 °C for 30 s, 55 °C for 30 s, 72 °C for


30 s, and a final extension at 72 °C for 5 min. The PCR products were visualized using 2% agarose gel electrophoresis. Dual index and Illumina sequencing adapters were joined via PCR using a


Nextera XT Index Kit. PCR product cleanup was performed using AMPure XP beads to purify the amplicons. The sizes of the PCR products were confirmed using a Bioanalyzer DNA 1000 Chip.


Library quantification was performed for quality control before sequencing using the Agilent Technologies 2100 Bioanalyzer. Pooled libraries were subjected to paired-end sequencing (2 × 300 


bp) using the Illumina MiSeq platform. Raw sequences were processed according to the QIIME2 pipeline [35]. Demultiplexed sequences from each sample were quality filtered and trimmed,


de-noised, and merged. The chimeric sequences were then identified and removed using the QIIME2 dada2 plugin. The QIIME2 feature-classifier plugin was then used to align amplicon sequence


variant (ASV) sequences against the SILVA database (version 132). The vegan package in R was used to calculate the alpha diversity, including the observed ASVs and Shannon and Simpson


indices. A principal coordinate analysis (PCoA) was performed using the Bray–Curtis distance. An analysis of variance (ANOVA) using distance matrices (Adonis) was performed to determine the


heterogeneity of fecal microbiota among the groups. A heatmap was plotted using the heatmap3 package in R. LIPOPOLYSACCHARIDE QUANTIFICATION Plasma and hepatic LPS levels were quantified


using a cell-based colorimetric assay for the detection of biologically active endotoxins. Plasma samples and liver homogenates (10 µL) were added to murine TLR4/NF-κB/secreted embryonic


alkaline phosphatase (SEAP) reporter HEK293 cells and incubated for 24 h. After incubation, the medium was removed, and the HEK-Blue™ LPS Detection Kit (InvivoGen, USA) was used to detect


TLR4 and an NF-κB-inducible SEAP. The abundances were then measured with a spectrophotometer at 620 nm [26]. INTESTINAL PERMEABILITY ANALYSIS Intestinal barrier permeability was examined by


gavage with fluorescein isothiocyanate-dextran (FITC-dextran; average molecular weight of 4,000; Sigma-Aldrich, 46944) as an intestinal permeability probe. Fifteen milligrams of FITC-dextran


in 0.2 mL of phosphate-buffered saline were gavaged to mice 3 h before sacrifice. Plasma FITC-dextran measurements were taken using a fluorometer at excitation and emission wavelengths of


485 and 538 nm (Fluoroskan Ascent FL, Thermo Fisher Scientific, USA), respectively, in black 96-well plates [36]. STATISTICAL ANALYSIS Data are presented as the mean ± standard deviation


(SD). A one-way ANOVA with Dunnett’s multiple comparison test was used to compare group means against the PL group. The Wilcoxon signed-rank test, Kruskal–Wallis test with Dunn’s multiple


comparison test, and ANOVA with Dunnett’s multiple comparison test were used to analyze the fecal microbiome dataset. All statistical analyses were performed using GraphPad Prism (version


9.1.2), R (version 3.6.1), or R Studio (version 1.2.5001). RESULTS CHEMICAL COMPOSITION OF GINGER ESSENTIAL OIL GEO was obtained by steam distillation, and the extraction rate was


approximately 0.14% (w/w). The gas chromatography (GC) analysis revealed that the GEO contained 18.8% neral and 12.2% geranial isomers (Supplementary Fig. 1). Both neral and geranial isomers


are geometric isomers of citral. Our results, therefore, confirmed that citral is the main component of GEO (approximately 31%). It was subsequently used for the animal studies. PALM


OIL-CONTAINING HFD WITH LPS INJECTION RESULTED IN AN ADVERSE EFFECT ON OBESOGENIC AND METABOLIC BIOMARKERS. SUPPLEMENTATION OF GEO AMELIORATE LIPIDEMIA The experimental design of this study


is shown in Fig. 1a. Eight-week-old C57BL/6 mice (_n_ = 7–8 per group) were fed a control diet or P-HFD with LPS injection (PL) with or without GEO supplementation via oral gavage. In this


study, we supplemented the mice daily with 12.5 (GL), 62.5 (GM), and 125 (GH) mg/kg for 12 weeks. The mice were sacrificed after 12 weeks to investigate the effect of GEO on the prevention


of obesogenic, metabolic, and hepatic changes. The PL group exhibited an increased body weight compared to the control (CON) group (_p_ = 0.0155) (Fig. 1b). We calculated the weight gain


rate based on the increase in body weight and time of feeding; the weight gain rate of the PL group was similar to the body weights, which were higher than those of the CON group (Fig. 1c).


At 12 weeks, the body weights in the PL group (30.8 ± 2.0) were significantly higher (_p_ = 0.0028) than those in the CON group (26.9 ± 0.8) (Fig. 1d). The relative total fat mass of the PL


group was found to be heavily increased compared to the control (_p_ < 0.0001), up to 195% of the CON group (Fig. 1e). The relative fat mass in different body locations in the PL group


was also higher than that in the CON group. This included the mass of the visceral, subcutaneous, epididymal white adipose tissue (WAT) (eWAT), inguinal WAT (iWAT), mesenteric WAT (mWAT),


perirenal WAT (pWAT), and inguinal subcutaneous white adipose tissue (isWAT), and interscapular brown adipose tissue (iBAT) (Fig. 1e, Supplementary Fig. 2). GEO supplementation, especially


at medium and high doses, reduced body weight changes (_p_ < 0.0001) (Fig. 1b) and tended to slightly lower final body weight and area under the curve of body weight (Fig. 1c, d). No


significant difference was observed in the average energy intake among the experimental groups, suggesting that diet type was the primary factor regulating body weight gain (Fig. 1f). The


study examined the plasma levels of glucose and lipidemic biomarkers, including fasting glucose, total triglyceride, total cholesterol, high-density lipoprotein cholesterol (HDL-C), and


low-density lipoprotein cholesterol (LDL-C) (Fig. 1g–k). Compared to the CON group, the PL group showed significant increases in plasma glucose (_p_ < 0.0001), total cholesterol (_p_ <


 0.0001), HDL-C (_p_ = 0.0004), and LDL-C (_p_ < 0.0001) levels, indicating the impact of PL on blood fasting glucose and lipid circulation. However, plasma total triglyceride levels were


reduced compared to the CON group. Supplementation with GEO at medium dosage reduced plasma glucose (_p_ = 0.0577) and total triglyceride (_p_ = 0.0125) levels compared to the PL group.


Additionally, low-dose GEO trended to improve plasma HDL-C levels (_p_ = 0.0666). Both medium and high doses of GEO significantly reduced plasma LDL-C levels (_p_ = 0.0312 and _p_ = 0.0001,


respectively). Collectively, supplementation of GEO, particularly at medium and high doses, significantly improves lipidemia. GEO PREVENTS NASH WITH REDUCED HEPATIC DAMAGE AND NAFLD ACTIVITY


SCORES AST and ALT are indicators of liver damage. We did not observe an increase in AST levels in the PL group compared to the CON group (Fig. 2a). The plasma ALT levels in the PL group


were markedly higher than those in the CON group (_p_ = 0.0208) (Fig. 2b). The plasma AST levels trended to be lower in the PL + GM and PL + GH groups. Remarkably, GEO supplementation in the


PL + GM and PL + GH groups significantly lowered the ALT levels compared to the PL group (_p_ = 0.0104 and _p_ = 0.0044, respectively). The liver weight in the PL group exhibited a


significant increase compared to the CON group (_p_ = 0.0006) (Fig. 2c). However, treatment with GEO did not lead to a reduction in liver weight. The representative image of the liver was


shown in Fig. 2d, and hepatomegaly was observed prominently in the PL group. We performed liver histological analyses to determine the impact of PL on NASH progression and the effect of GEO


on NASH prevention. Hematoxylin and eosin (H&E) staining of the liver sections showed that the lipid droplets in the PL group were numerous compared to those in the CON group. Based on


the NAS, mouse liver sections were examined for steatosis, hepatocyte ballooning, and lobular inflammation scores (Fig. 2d–h). A NAS ≥ 5 was considered to be indicative of NASH. PL induced


the NASH phenotype by increasing the NAS (median = 5) compared to the CON group (median = 0) (_p_ < 0.0001), resulting in a prevalence of 85.7% for NASH. GEO at low, medium, and high


dosages reduced the prevalence of NASH to 50%, 0%, and 0%, respectively, with respective NAS of 4.5, 4, and 3.5. PL-treated mice exhibited increased levels and prevalence of hepatic


steatosis, hepatocyte ballooning, and lobular inflammation. GEO exhibited a potential effect on NASH prevention by reversing these scores. The PL + GM and PL + GH groups showed a reduction


in the hepatocyte ballooning score compared with the PL group (_p_ = 0.0499). Based on the histopathological analysis of liver biopsy results, it was evident that GEO supplementation led to


an overall improvement in liver inflammation. The primary mechanism by which GEO reduced the severity of NASH was primarily through the reduction of hepatocyte ballooning. Medium and high


doses of GEO were more effective for NASH prevention in PL-treated mice. Overall, these data suggest that GEO can potentially prevent NASH. GEO IMPROVES HEPATIC ANTIOXIDANT ENZYME ACTIVITIES


A HFD increases fat uptake, and excess fat can accumulate in the liver by increasing ROS and lipid peroxidation, which further causes liver cell damage. Several hepatic antioxidants and


enzymes have been found to eliminate ROS. CYP2E1 further increases hepatic oxidative stress. Our data revealed that the PL group exhibited increased CYP2E1 protein expression (Fig. 3a). GEO


supplementation trended to reduce CYP2E1 expression. In NASH patients, impaired glutathione metabolism and reduced enzyme activities in the blood support a consistent role of free radical


cytotoxicity in the pathophysiology of the disease [37]. PL resulted in the reduction of SOD and GSH levels. Interestingly, GEO supplementation enhanced CAT, GSH, and GRd levels. GPx trended


to increase in the GEO-supplemented group (Fig. 3b–f). The overall antioxidant capacity of a high dose of GEO (GH) was more beneficial than that of low and medium doses of GEO. GEO may,


therefore, suppress oxidative stress in PL-induced NASH by improving antioxidant and antioxidant enzyme activities. GEO DECREASES HEPATIC INFLAMMATION BY SUPPRESSING PRO-INFLAMMATORY


CYTOKINES AND MEDIATING THE NLRP3/ACS/CASPASE-1 PATHWAY According to the histopathological analysis of the liver biopsy results, GEO alleviated the NAS mainly by reversing steatosis and the


hepatocyte ballooning score. We measured the levels of hepatic pro-inflammatory cytokines. PL induced an increase in TNF-α, IL-1β, and IL-6 levels. GEO at medium and high doses significantly


reduced hepatic IL-1β (_p_ = 0.0013 and _p_ < 0.0001, respectively) and IL-6 levels (_p_ < 0.0001 and _p_ < 0.0001, respectively). In addition, a high dose of GEO significantly


decreased TNF-α levels (_p_ = 0.0012) (Fig. 4a–c). To investigate the role of GEO in hepatic fatty acid synthesis and β-oxidation, we measured SREBP-1, PPARα, and HMGCR protein expression.


PL increased SREBP-1 and HMGCR levels. GEO further favored the reduction of these biomarkers. An increase in PPARα expression was also observed in the GEO supplemented group (Supplementary


Fig. 3). Although the hepatic lipid regulation-related protein expression did not show a significant difference, alteration trends were observed, suggesting that GEO partially exerts a


beneficial effect against fatty synthesis and lipid oxidation to prevent NASH. Several studies have further suggested that the NLRP3 inflammasome plays a crucial role in NASH progression.


Therefore, we investigated the expression of NLRP3 and its downstream proteins in the liver. The hepatic NLRP3, ACS, and caspase-1 protein levels are shown in Fig. 4d. PL intervention for 12


weeks induced an increase in hepatic NLRP3 and ASC expression compared to control mice (_p_ = 0.0012 and _p_ = 0.0011, respectively) (Fig. 4e–f). All doses of GEO significantly suppressed


NLRP3 and ASC protein expression compared to the PL group (_p_ < 0.05). In addition, the hepatic protein expression of caspase-1 trended to increase in the PL group, but was reduced in


the GEO-supplemented group (Fig. 4g). These results suggest that GEO mediates the NLRP3/ASC/caspase-1 pathway, resulting in the reduction of pro-inflammatory cytokine production to prevent


NASH. PL AND GEO REMODEL THE FECAL MICROBIOTA COMPOSITION, AND GEO REVERSES GUT MICROBIOTA DYSBIOSIS THAT MAY BENEFIT NASH AMELIORATION The gut microbiota plays an important role in the


pathogenesis of NASH and its metabolites. To understand the effect of PL and GEO, we sequenced the gene encoding the bacterial V3–V4 16 S rRNA amplicons obtained from feces sampled from the


colon. The amplicon sequence variant (ASV) table was generated using the QIIME2 pipeline against the SILVA database (version 132). A total of 1093 ASVs were obtained and assigned to


taxonomies at the species level (140 species). We calculated the α-diversity based on the observed ASVs and Shannon and Simpson indices from the ASV table. The observed ASV of the PL group


trended to decrease, but the Shannon and Simpson diversity indices significantly increased (Fig. 5a). GEO did not cause a shift in α-diversity. The β-diversity based on the Bray−Curtis


distance (PC1: 35.95% and PC2: 11.31%) demonstrated that both PL intervention and GEO significantly impact gut microbiota remodeling (Adonis, _p_ < 0.001). PL intervention was a primary


factor in the shift in the gut microbiota in PCoA1 (Fig. 5b). GEO further played a secondary role in gut microbiota remodeling in a dose-dependent manner in PCoA2. Among the GEO-treated


groups, the gut microbiota shifted from that of the PL group in a dose-dependent manner. A high dose of GEO (PL + GH group) shifted outward from the PL group, suggesting that a higher dosage


of GEO exhibits a more substantial effect on gut microbiota remodeling. We further assessed the association between NASH-related features and gut microbiota composition using the envfit


function of the vegan R package. The results are shown in Fig. 5b as red vectors. The direction of the vectors from the centroid was opposite to that of the control group and encountered


NASH-phenotype mice. NASH- and obesity-related parameters exhibited a significant association with the gut microbiota of NASH mice (_p_ < 0.05), including NASH activity scores, hepatocyte


ballooning, steatosis scores, body weights, LDLs, TNF-α, and IL-1β. The heatmap showed 42 different significant genera based on the Kruskal–Wallis test with an FDR-adjusted _p_-value (_p_ 


< 0.05), as presented in Fig. 5c. Heatmap hierarchical clustering displayed two main clusters, indicating that PL intervention was the main factor that altered the gut microbiota at the


genus level. Spearman’s correlation analysis was then performed to assess NASH and obesity biomarkers. The results demonstrated that 16 genera were positively correlated with NASH and


obesity biomarkers; conversely, 12 genera were negatively correlated (Fig. 5d). Among them, the NASH-related microbiota, including _Tyzzerella_ and _Blautia_, showed a positive correlation.


In contrast, beneficial bacteria, such as _Lactobacillus_, _Alistipes_, _Olsenella_, and _Bifidobacterium_, exhibited a negative correlation. However, the correlation between GEO and


NASH-related microbiota was reduced, suggesting that GEO supplementation may protect against the dysbiosis of gut microbiota. The NASH-related microbiota, including _Blautia_ and


_Tyzzerella_, trended to be suppressed by GEO, especially at high doses (_p_ = 0.1216 and _p_. = 0.0756, respectively) (Supplementary Fig. 4a, b). Beneficial genera, including _Alitipes_,


_Bifidobacterium_, _Lactobacillus_, and _Olsenella_, were depleted in the PL group. GEO at a high dose trended to recover _Alistpes_ and _Lactobacillus_, and the low dose resulted in the


enrichment of _Olsenella_ (Supplementary Fig. 4c–f). GEO supplementation in PL may therefore reverse gut microbiota dysbiosis and ameliorate NASH. GEO ALLEVIATES NASH BY MODULATING THE


LPS/TLR4/NF-ΚB SIGNALING PATHWAY Unhealthy diets, low in fiber and high in fat and sugar, can lead to gut microbiota dysbiosis and intestinal barrier leakage. This compromised barrier


facilitates the translocation of gut microbiota-derived metabolites, such as LPS, into the liver [13, 14]. GEO resulted in favorable gut microbiota remodeling, as described in the previous


section. To understand the beneficial effect of GEO on intestinal permeability, we administered fluorescein isothiocyanate (FITC)-dextran to the mice and detected the fluorescence intensity


in the plasma. PL trended to increase the fluorescence intensity in the plasma, suggesting that PL intervention results in intestinal leakage. GEO supplementation trended to reduce


intestinal permeability (Fig. 6a). The dysfunction of the intestinal barrier allows gut microbial LPS to enter the liver and circulatory system through the portal vein and lymphatic system,


thereby promoting inflammation. The murine TLR4/NF-κB/SEAP reporter HEK293 cell assay was used to detect LPS levels. The results demonstrated no significant differences in plasma LPS levels


among the control, PL-, and GEO-treated groups (Fig. 6b). Interestingly, hepatic LPS levels were markedly elevated (_p_ < 0.0001) compared to those in the CON group. GEO supplementation


in PL intervention at low, medium, and high doses significantly reduced hepatic LPS levels (_p_ < 0.0001, _p_ = 0.0001, and _p_ < 0.0001, respectively) (Fig. 6c). Mechanistically,


gut-derived LPS activated the TLR4/NF-κB pathway and triggered hepatic inflammation. We further examined the hepatic protein expression of TLR4 and NF-κB. Both hepatic TLR4 and NF-κB levels


were higher in the PL group than in the CON group (_p_ = 0.0141 and _p_ = 0.0022, respectively) (Fig. 6d, e). Low, medium, and high doses of GEO trended to reduced TLR4 (_p_ = 0.1142, _p_ = 


0.0448, and _p_ = 0.0029, respectively) and NF-κB protein expression (_p_ = 0.0053, _p_ = 0.0025, and _p_ < 0.0001, respectively) (Fig. 6e, f), thereby resulting in the reduction of the


pro-inflammatory cytokines TNF-α, IL-1β, and IL-6, and the NAS (Fig. 4a–c and Fig. 2e). Collectively, these results demonstrate the potential protective effect of GEO against NASH via


modulating the gut microbiota and its metabolite–liver axis through the LPS/TLR4/NF-κB signaling pathway. DISCUSSION A trans-fat-containing HFD is an excellent model for inducing NASH in


animals [24]. Due to the prohibition of using trans-fats in the food supply chain and food industry [23], however, trans-fat-containing HFDs may not be suitable for mimicking NASH


progression in humans. The replacement of trans-fat with palm oil in a HFD exhibits advantages and the potential for inducing NASH with human NASH phenotype translatability [25]. P-HFD-fed


rodents manifest morphological features analogous to those of human NASH, including macrosteatosis, lobular inflammation, hepatocyte ballooning degeneration, and periportal/perisinusoidal


fibrosis [25]. Furthermore, we developed a P-HFD supplemented with an intraperitoneal injection of LPS (PL) to mimic the conditions of gut dysbiosis and endotoxemia. PL causes NASH,


intestinal leakage, endotoxemia, and dysbiosis of the intestinal microbiota by increasing pathogenic bacteria and reducing beneficial microbiota [26]. Therefore, a PL mouse model was used in


this study. Our results demonstrated that PL intervention for 12 weeks negatively affected obesogenic and metabolic biomarkers in mice, including body weight, the area under the curve (AUC)


of body weight gain, total fat mass, plasma glucose, total cholesterol, and LDL-C (Fig. 1). The average energy intake of the mice among the groups was not significantly different,


suggesting that the adverse obesogenic and metabolic consequences were due to the P-HFDs and LPS injections. The P-HFD diet comprised a large amount of palm oil, fat, fructose, and


cholesterol. A previous study found that dietary saturated fat and fructose intake are associated with intrahepatic lipid accumulation, lipogenesis, insulin resistance, oxidative stress, and


inflammation [38, 39]. Palm oil contains a large amount of saturated fatty palmitic acid (C16:0) [40]. Palm oil in normocaloric and normolipidic diets can adversely interfere with and


exacerbate metabolic and glucose homeostasis and induce inflammation in the liver and white adipose tissue. The consequence is more acute in mice administered interesterified palm oil [41].


Injections of LPS in choline-deficient l-amino-acid-defined (CDAA)-fed mice for 16 weeks have been found to intensify hepatic inflammation and pericellular fibrosis via the TLR4/NF-κB


signaling pathway [42]. This study demonstrated that the use of PL could be more advantageous than the CDAA diet as PL can mimic the human diet and endotoxemia, whereas the CDAA diet is not


practical in humans. In this study, the level of plasma TG declined in the PL group compared to the CON group, which is consistent with previous studies [24, 25] and was likely due to blood


cholesterol in the P-HFD inhibiting lipoprotein synthesis, thereby reducing triglyceride secretion from the liver to the circulation [43, 44]. In this study, GEO supplementation prevented


the obesogenic metabolic phenotype and ameliorated lipidemia by reducing the body weight and plasma glucose, total triglycerides, and LDL-C, and by increasing HDL-C. GEO further alleviated


the NASH phenotype by attenuating the plasma hepatic damage biomarkers AST, ALT, and NAS, mainly by reducing the hepatocyte ballooning score. Hepatocyte ballooning is generally considered to


be a form of apoptosis related to fibrosis and cytoskeletal damage [45]. In this study, medium and high doses of GEO (62.5 and 125 mg/kg bw) exhibited a more substantial hepatoprotective


effect. GEO displays an anti-hyperlipidemic effect by reducing serum FFA, triglycerides, and total cholesterol levels in mice, previously reported [6]. GEO exhibits hepatoprotective


properties against alcoholic fatty liver disease and can change particular metabolites in the blood [32]. HFD intake is associated with increased oxidative stress in the liver, which is one


of the pathogeneses of NASH [46]. NASH patients have been found to exhibit increased CYP2E1, which promotes ROS generation and hepatic oxidative stress [47]. Several studies have further


demonstrated that ginger exerts antioxidant activity [6, 32, 48]. In this study, GEO supplementation trended to lower hepatic CYP2E1 protein expression and significantly enhanced the hepatic


antioxidant enzyme capacity, including CAT, GRd, and GSH. These results demonstrate that GEO alleviates hepatic oxidative stress in mice with PL-induced NASH. HFD consumption interferes


with homeostasis by impairing lipid metabolism, resulting in imbalanced lipid uptake and storage and leading to hepatic steatosis [7, 49]. The PL-induced NASH mouse group showed a trended to


increase in the hepatic protein expression of SREBP-1 and HMGCR compared to the control mice. Both of these transcription factors play essential roles in the regulation of hepatic lipid


biosynthesis. The elevation of SREBP-1c promotes triglyceride synthesis, and HMGCR is a key enzyme in cholesterol synthesis [50]. In addition, PPARα, a regulator of fatty acid β-oxidation,


decreased in PL-induced NASH mice. The effect of GEO on these lipid-regulating proteins exhibited a substantial improve, but it did not reach statistical significance, which was consistent


with the histopathological data. The different forms of diet-induced NASH models could induce different degrees of NASH severity. PL-induced NASH showed substantial severity compared to our


previous study, in which HFD induced simple hepatic steatosis [6, 51]. GEO alleviated the NASH phenotype and pro-inflammatory cytokines TNF-α, IL-6, and IL-1β. Therefore, we investigated the


underlying mechanisms of inflammation. The NLRP3 inflammasome plays an essential role in the hepatic inflammatory response [52]. NASH patients and animal models have exhibited increases in


the activation of the NLRP3 inflammasome [17, 52]. Blocking the NLRP3 inflammasome attenuates hepatic steatosis, inflammation, and fibrosis in mice [17]. In our study, the expression levels


of NLRP3 and ASC were increased in PL-induced mice, whereas they were reduced with GEO supplementation. Caspase-1 protein expression was trended to suppressed by GEO. The activation of the


NLRP3 inflammasome is stimulated by danger-associated molecular patterns and PAMPs such as LPS [53]. The LPS-mediated TLR4/NF-κB pathway can activate the NLRP3 inflammasome by enhancing the


expression of NLRP3 [54]. In in vitro experiments, geranial and neral isomers of citral in GEO suppressed the production of the NLRP3 inflammasome and pro-IL-1β [55]. Therefore, GEO


exhibited anti-inflammatory activity by reducing NLRP3 inflammasome activation, which may be caused by citral and its isomers. Diet is a primary factor in modifying the composition of gut


microbiota [56]. PL intervention is a prime factor in modifying the fecal microbiota composition of mice. GEO has less influence on α-diversity. Similarly, PCoA β-diversity based revealed


that diet is the principal factor that shapes the gut microbiota. GEO plays a secondary role in modifying gut microbiota bacterial diversity. A high dose of GEO resulted in the gut


microbiota shifting outward from the PL-treated group. GEO reduced the relative abundance of NASH-associated bacteria, including _Blautia_ and _Tyzzerella_, which are considered potentially


harmful. Compared to healthy individuals, _Blautia_ abundance increased in NASH patients and was positively correlated with LPS levels [57]. _Tyzzerella_ has been reported to be enriched


after 12 weeks of HFD intervention and is positively associated with liver dysfunction-associated parameters [58]. In contrast, GEO supplementation in PL mice enhanced the relative abundance


of _Lactobacillus_, _Alistipes_, and _Olsenella_, which are considered beneficial bacteria. A previous study reported a lower relative abundance of _Lactobacillus_ in GAN _ob/ob_-NASH mice


[24]. _Lactobacillus_ may be used as a probiotic to improve steatohepatitis through the gut microbiota–liver axis by modulating gut microbiota composition and the inflammatory pathway in


NAFLD [59]. _Alistipes_, which are potential SCFA-producing bacteria that reduce hepatic fibrotic conditions through cytokine modulation, have been found at lower levels in the guts of NASH


and NASH-cirrhosis patients [60, 61]. _Olsenella_, an SCFA producer, is associated with tight junction improvement [62, 63]. Thus, GEO supplementation can remodel gut microbiota composition


and reverse gut microbiota dysbiosis, which may contribute to ameliorating NASH. This study has a limitation in that all mice for each treatment were housed in separate cages, and the mice


did not receive the same microbiomes from the start. Given that mice are coprophagic, this may introduce a potential cage effect. To mitigate these confounding factors, future studies should


consider co-housing the mice and ensuring they receive the same microbiomes from the beginning. This approach would allow for a more precise assessment of the effects of diet and dietary


supplements on the gut microbiota. The elevated localization of LPS in hepatocytes has been reported in NAFLD and may cause liver inflammation via a TLR4-related pathway [64]. This study


found no significant changes in intestinal permeability or plasma LPS levels in PL-induced mice, whereas hepatic LPS levels were noted to increase. Although we observed increased intestinal


permeability of PL mice, the lack of statistical significance may be due to individual variations in gut permeability within the PL group. Based on our experimental design, we administered


P-HFD mice with weekly intraperitoneal injections of LPS. The low-dose, weekly intraperitoneal injection of LPS in our study is unlikely to have substantially affected plasma and hepatic LPS


levels, as the observed levels were relatively low compared to the injection dose. Additionally, LPS in the circulation was found at a level similar to that in the control group, these data


suggested that the elevation of hepatic LPS levels in the liver was derived from the gut and not the extra injection of LPS. LPS is a risk factor for inducing hepatic inflammation and NASH.


The upregulation of the LPS-TLR4 pathway leads to NF-κB activation and inflammatory cytokine production, which play key roles in NASH progression and development [42]. PL-induced NASH mice


exhibited a higher expression of TLR4 and NF-κB, which resulted in a more severe inflammatory condition than mice supplemented with GEO. In addition to the TLR4/NF-κB pathway, LPS is also


involved in activating the NLRP3 inflammasome [65]. We also conducted a dosage translation of GEO from mice to humans and compared it with the dosage achievable from natural ginger


(Supplementary Fig. 5). Our analysis indicates that the dosage used in the study can be obtained from natural ginger, suggesting the potential development of GEO as a functional food or food


supplement. Nevertheless, further investigation through human studies is required to comprehensively evaluate its potential applications. Histological analysis of kidney, spleen, and ileum


sections in the GEO-treated group did not reveal any significant findings, as shown in Supplementary Fig. 6. These results indicate the safety of GEO under our experimental conditions.


Moreover, a subchronic toxicity study conducted on Wistar rats for a duration of 13 weeks reported no observed adverse effect level (NOAEL) of 500 mg/kg body weight [66]. When extrapolated


to mouse dosage [67], this corresponds to 1000 mg/kg body weight. Importantly, it should be noted that the dosages utilized in our study were relatively lower than the NOAEL reported in the


literature, providing further evidence of the safety profile of GEO at the selected dosages. Collectively, the results of our study demonstrated that GEO exhibits a potential


hepatoprotective effect by preventing NASH progression in a PL-induced NASH mouse model. The mechanisms of action of GEO may include reducing liver steatosis and oxidative stress, blocking


the NLRP3 inflammasome pathway, and remodeling the LPS/TLR4/NF-κB pathway which could be modulated by changes in gut microbiota. GEO, therefore, may be used as a dietary supplement to


prevent NASH. DATA AVAILABILITY The raw 16 s rRNA sequencing data used to produce all figures are accessible in the NCBI Short Read Archive under the following accession numbers: BioProject


(PRJNA854318), BioSample (SAMN29428094), and SRA (SRR19905849-83). CODE AVAILABILITY Bioinformatic tools, software version, parameters, and open-source code used in this present study are


described in the “Methods” section. More details regarding the code to reproduce the analyses are available upon request. REFERENCES * Younossi Z, Anstee QM, Marietti M, Hardy T, Henry L,


Eslam M, et al. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nat Rev Gastroenterol Hepatol. 2018;15:11–20. Article  PubMed  Google Scholar  * Brunt EM,


Wong VW, Nobili V, Day CP, Sookoian S, Maher JJ, et al. Nonalcoholic fatty liver disease. Nat Rev Dis Primers. 2015;1:15080. Article  PubMed  Google Scholar  * Aron-Wisnewsky J, Vigliotti C,


Witjes J, Le P, Holleboom AG, Verheij J, et al. Gut microbiota and human NAFLD: disentangling microbial signatures from metabolic disorders. Nat Rev Gastroenterol Hepatol. 2020;17:279–97.


Article  PubMed  Google Scholar  * Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism. 2016;65:1038–48. Article  CAS


  PubMed  Google Scholar  * Arroyave-Ospina JC, Wu ZM, Geng YN, Moshage H. Role of oxidative stress in the pathogenesis of non-alcoholic fatty liver disease: Implications for prevention and


therapy. Antioxidants. 2021;10:174. Article  CAS  PubMed  PubMed Central  Google Scholar  * Lai YS, Lee WC, Lin YE, Ho CT, Lu KH, Lin SH, et al. Ginger Essential oil ameliorates hepatic


injury and lipid accumulation in high fat diet-induced nonalcoholic fatty liver disease. J Agric Food Chem. 2016;64:2062–71. Article  CAS  PubMed  Google Scholar  * Ipsen DH, Lykkesfeldt J,


Tveden-Nyborg P. Molecular mechanisms of hepatic lipid accumulation in non-alcoholic fatty liver disease. Cell Mol Life Sci. 2018;75:3313–27. Article  CAS  PubMed  PubMed Central  Google


Scholar  * Han KH, Hashimoto N, Fukushima M. Relationships among alcoholic liver disease, antioxidants, and antioxidant enzymes. World J Gastroentero. 2016;22:37–49. Article  CAS  Google


Scholar  * Swanson KV, Deng M, Ting JP. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol. 2019;19:477–89. Article  CAS  PubMed  PubMed Central 


Google Scholar  * Vandanmagsar B, Youm YH, Ravussin A, Galgani JE, Stadler K, Mynatt RL, et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat


Med. 2011;17:179–88. Article  CAS  PubMed  PubMed Central  Google Scholar  * Gentile CL, Weir TL. The gut microbiota at the intersection of diet and human health. Science. 2018;362:776–80.


Article  CAS  PubMed  Google Scholar  * Jennison E, Byrne CD. The role of the gut microbiome and diet in the pathogenesis of non-alcoholic fatty liver disease. Clin Mol Hepatol.


2021;27:22–43. Article  PubMed  Google Scholar  * Hrncir T, Hrncirova L, Kverka M, Hromadka R, Machova V, Trckova E, et al. Gut microbiota and NAFLD: pathogenetic mechanisms, microbiota


signatures, and therapeutic interventions. Microorganisms. 2021;9:957. Article  CAS  PubMed  PubMed Central  Google Scholar  * Albillos A, de Gottardi A, Rescigno M. The gut-liver axis in


liver disease: Pathophysiological basis for therapy. J Hepatol. 2020;72:558–77. Article  CAS  PubMed  Google Scholar  * Dapito DH, Mencin A, Gwak GY, Pradere JP, Jang MK, Mederacke I, et al.


Promotion of hepatocellular carcinoma by the intestinal microbiota and TLR4. Cancer Cell. 2012;21:504–16. Article  CAS  PubMed  PubMed Central  Google Scholar  * Kolodziejczyk AA, Zheng DP,


Shibolet O, Elinav E. The role of the microbiome in NAFLD and NASH. Embo Mol Med. 2019;11:e9302. Article  PubMed  Google Scholar  * Wree A, McGeough MD, Pena CA, Schlattjan M, Li HY,


Inzaugarat ME, et al. NLRP3 inflammasome activation is required for fibrosis development in NAFLD. J Mol Med. 2014;92:1069–82. Article  CAS  PubMed  Google Scholar  * Ikawa-Yoshida A, Matsuo


S, Kato A, Ohmori Y, Higashida A, Kaneko E, et al. Hepatocellular carcinoma in a mouse model fed a choline-deficient, L-amino acid-defined, high-fat diet. Int J Exp Pathol. 2017;98:221–33.


Article  CAS  PubMed  PubMed Central  Google Scholar  * Mu YP, Ogawa T, Kawada N. Reversibility of fibrosis, inflammation, and endoplasmic reticulum stress in the liver of rats fed a


methionine-choline-deficient diet. Lab Invest. 2010;90:245–56. Article  CAS  PubMed  Google Scholar  * Clapper JR, Hendricks MD, Gu G, Wittmer C, Dolman CS, Herich J, et al. Diet-induced


mouse model of fatty liver disease and nonalcoholic steatohepatitis reflecting clinical disease progression and methods of assessment. Am J Physiol Gastrointest Liver Physiol.


2013;305:G483–95. Article  CAS  PubMed  Google Scholar  * Brownell KD, Pomeranz JL. The trans-fat ban-food regulation and long-term health. N Engl J Med. 2014;370:1773–5. Article  CAS 


PubMed  Google Scholar  * Grossman MR. USA: FDA issues order to ban artificial trans fat by 2018 reports. Eur Food Feed L Rev. 2015;10:317. Google Scholar  * Parziale A, Ooms G. The global


fight against trans-fat: the potential role of international trade and law. Global Health. 2019;15:46. Article  PubMed  PubMed Central  Google Scholar  * Boland ML, Oro D, Tolbol KS, Thrane


ST, Nielsen JC, Cohen TS, et al. Towards a standard diet-induced and biopsy-confirmed mouse model of non-alcoholic steatohepatitis: impact of dietary fat source. World J Gastroenterol.


2019;25:4904–20. Article  CAS  PubMed  PubMed Central  Google Scholar  * Hansen HH, Ægidius HM, Oro D, Evers SS, Heeboll S, Eriksen PL, et al. Human translatability of the GAN diet-induced


obese mouse model of non-alcoholic steatohepatitis. BMC Gastroenterol. 2020;20:210. Article  CAS  PubMed  PubMed Central  Google Scholar  * Sheen LY, Panyod S, Wu WK, Hsieh YC, Tseng YJ,


Peng SY, et al. Establishment of animal models for NASH and utilization for mining associated gut microbiota. Res Sq. 2022. https://doi.org/10.21203/rs.3.rs-1826044/v1. * Pappachan JM, Babu


S, Krishnan B, Ravindran NC. Non-alcoholic fatty liver disease: a clinical update. J Clin Transl Hepatol. 2017;5:384–93. PubMed  PubMed Central  Google Scholar  * Ganesan K, Jayachandran M,


Xu B. A critical review on hepatoprotective effects of bioactive food components. Crit Rev Food Sci Nutr. 2018;58:1165–229. Article  CAS  PubMed  Google Scholar  * Wang X, Shen Y, Thakur K,


Han J, Zhang JG, Hu F, et al. Antibacterial activity and mechanism of ginger essential oil against _Escherichia coli_ and _Staphylococcus aureus_. Molecules. 2020;25:3955. Article  CAS 


PubMed  PubMed Central  Google Scholar  * Nerilo SB, Rocha GHO, Tomoike C, Mossini SAG, Grespan R, Mikcha JMG, et al. Antifungal properties and inhibitory effects upon aflatoxin production


by Zingiber officinale essential oil in Aspergillus flavus. Int J Food Sci Tech. 2016;51:286–92. Article  CAS  Google Scholar  * Panyod S, Wu WK, Peng SY, Tseng YJ, Hsieh YC, Chen RA, et al.


Ginger essential oil and citral ameliorates atherosclerosis in ApoE−/− mice by modulating trimethylamine-N-oxide and gut microbiota. NPJ Sci Food. 2023;7:19. Article  PubMed  PubMed Central


  Google Scholar  * Liu CT, Raghu R, Lin SH, Wang SY, Kuo CH, Tseng YJ, et al. Metabolomics of ginger essential oil against alcoholic fatty liver in mice. J Agric Food Chem.


2013;61:11231–40. Article  CAS  PubMed  Google Scholar  * Liang W, Menke AL, Driessen A, Koek GH, Lindeman JH, Stoop R, et al. Establishment of a general NAFLD scoring system for rodent


models and comparison to human liver pathology. PLoS ONE. 2014;9:e115922. Article  PubMed  PubMed Central  Google Scholar  * Panyod S, Wu WK, Ho CT, Lu KH, Liu CT, Chu YL, et al. Diet


supplementation with allicin protects against alcoholic fatty liver disease in mice by improving anti-inflammation and antioxidative functions. J Agric Food Chem. 2016;64:7104–13. Article 


CAS  PubMed  Google Scholar  * Bolyen E, Rideout JR, Dillon MR, Bokulich NA, Abnet CC, Al-Ghalith GA, et al. Reproducible, interactive, scalable and extensible microbiome data science using


QIIME 2 (vol 37, pg 852, 2019). Nat Biotechnol. 2019;37:1091. Article  CAS  PubMed  Google Scholar  * Chassaing B, Koren O, Goodrich JK, Poole AC, Srinivasan S, Ley RE, et al. Dietary


emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome. Nature. 2015;519:92–6. Article  CAS  PubMed  PubMed Central  Google Scholar  * Nobili V, Pastore A,


Gaeta LM, Tozzi G, Comparcola D, Sartorelli MR, et al. Glutathione metabolism and antioxidant enzymes in patients affected by nonalcoholic steatohepatitis. Clin Chim Acta. 2005;355:105–11.


Article  CAS  PubMed  Google Scholar  * Lim JS, Mietus-Snyder M, Valente A, Schwarz JM, Lustig RH. The role of fructose in the pathogenesis of NAFLD and the metabolic syndrome. Nat Rev


Gastro Hepat. 2010;7:251–64. Article  CAS  Google Scholar  * Meex RCR, Blaak EE. Mitochondrial dysfunction is a key pathway that links saturated fat intake to the development and progression


of NAFLD. Mol Nutr Food Res. 2021;65:e1900942. Article  PubMed  Google Scholar  * Montoya C, Cochard B, Flori A, Cros D, Lopes R, Cuellar T, et al. Genetic architecture of palm oil fatty


acid composition in cultivated oil palm (_Elaeis guineensis_ Jacq.) compared to its wild relative E. Oleifera (HBK) Cortes. PLoS ONE. 2014;9:e95412. Article  PubMed  PubMed Central  Google


Scholar  * Miyamoto JE, Reginato A, Portovedo M, dos Santos RM, Stahl MA, Le Stunff H, et al. Interesterified palm oil impairs glucose homeostasis and induces deleterious effects in liver of


Swiss mice. Metab Clin Exp. 2020;112:154350. Article  CAS  PubMed  Google Scholar  * Nakanishi K, Kaji K, Kitade M, Kubo T, Furukawa M, Saikawa S, et al. Exogenous administration of


low-dose lipopolysaccharide potentiates liver fibrosis in a choline-deficient l-amino-acid-defined diet-induced murine steatohepatitis model. Int J Mol Sci. 2019;20:2724. Article  CAS 


PubMed  PubMed Central  Google Scholar  * Ma K, Malhotra P, Soni V, Hedroug O, Annaba F, Dudeja A, et al. Overactivation of intestinal SREBP2 in mice increases serum cholesterol. PLoS ONE.


2014;9:e84221. Article  PubMed  PubMed Central  Google Scholar  * Henkel J, Coleman CD, Schraplau A, Jhrens K, Weber D, Castro JP, et al. Induction of steatohepatitis (NASH) with insulin


resistance in wildtype B6 mice by a western-type diet containing soybean oil and cholesterol. Mol Med. 2017;23:70–82. Article  CAS  PubMed  PubMed Central  Google Scholar  * Caldwell S,


Ikura Y, Dias D, Isomoto K, Yabu A, Moskaluk C, et al. Hepatocellular ballooning in NASH. J Hepatol. 2010;53:719–23. Article  PubMed  PubMed Central  Google Scholar  * Dhibi M, Brahmi F,


Mnari A, Houas Z, Chargui I, Bchir L, et al. The intake of high fat diet with different trans fatty acid levels differentially induces oxidative stress and non alcoholic fatty liver disease


(NAFLD) in rats. Nutr Metab. 2011;8:65. Article  CAS  Google Scholar  * Aubert J, Begriche K, Knockaert L, Robin MA, Fromenty B. Increased expression of cytochrome P450 2E1 in nonalcoholic


fatty liver disease: mechanisms and pathophysiological role. Clin Res Hepatol Gastroenterol. 2011;35:630–7. Article  CAS  PubMed  Google Scholar  * Stoilova I, Krastanov A, Stoyanova A,


Denev P, Gargova S. Antioxidant activity of a ginger extract (Zingiber officinale). Food Chem. 2007;102:764–70. Article  CAS  Google Scholar  * Chen Z, Tian R, She Z, Cai J, Li H. Role of


oxidative stress in the pathogenesis of nonalcoholic fatty liver disease. Free Radic Biol Med. 2020;152:116–41. Article  CAS  PubMed  Google Scholar  * Sekiya M, Yahagi N, Matsuzaka T,


Takeuchi Y, Nakagawa Y, Takahashi H, et al. SREBP-1-independent regulation of lipogenic gene expression in adipocytes. J Lipid Res. 2007;48:1581–91. Article  CAS  PubMed  Google Scholar  *


Lai YS, Chen WC, Ho CT, Lu KH, Lin SH, Tseng HC, et al. Garlic essential oil protects against obesity-triggered nonalcoholic fatty liver disease through modulation of lipid metabolism and


oxidative stress. J Agric Food Chem. 2014;62:5897–906. Article  CAS  PubMed  Google Scholar  * Thomas H. NAFLD: A critical role for the NLRP3 inflammasome in NASH. Nat Rev Gastroenterol


Hepatol. 2017;14:197. Article  CAS  PubMed  Google Scholar  * Gong T, Liu L, Jiang W, Zhou R. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat Rev Immunol.


2020;20:95–112. Article  CAS  PubMed  Google Scholar  * Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med. 2015;21:677–87. Article 


PubMed  PubMed Central  Google Scholar  * Liao PC, Yang TS, Chou JC, Chen J, Lee SC, Kuo YH, et al. Anti-inflammatory activity of neral and geranial isolated from fruits of _Litsea cubeba_


Lour. J Funct Foods. 2015;19:248–58. Article  CAS  Google Scholar  * Leeming ER, Johnson AJ, Spector TD, Le Roy CI. Effect of diet on the gut microbiota: Rethinking intervention duration.


Nutrients. 2019;11:2862. Article  PubMed  PubMed Central  Google Scholar  * Del Chierico F, Nobili V, Vernocchi P, Russo A, De Stefanis C, Gnani D, et al. Gut microbiota profiling of


pediatric nonalcoholic fatty liver disease and obese patients unveiled by an integrated meta-omics-based approach. Hepatology. 2017;65:451–64. Article  PubMed  Google Scholar  * Li RJ, Jie


ZY, Feng Q, Fang RL, Li F, Gao Y, et al. Network of interactions between gut microbiome, host biomarkers, and urine metabolome in carotid atherosclerosis. Front Cell Infect Mi.


2021;11:708088. Article  CAS  Google Scholar  * Lee NY, Yoon SJ, Han DH, Gupta H, Youn GS, Shin MJ, et al. _Lactobacillus_ and _Pediococcus_ ameliorate progression of non-alcoholic fatty


liver disease through modulation of the gut microbiome. Gut Microbes. 2020;11:882–99. Article  PubMed  PubMed Central  Google Scholar  * Astbury S, Atallah E, Vijay A, Aithal GP, Grove JI,


Valdes AM. Lower gut microbiome diversity and higher abundance of proinflammatory genus _Collinsella_ are associated with biopsy-proven nonalcoholic steatohepatitis. Gut Microbes.


2020;11:569–80. Article  PubMed  Google Scholar  * Parker BJ, Wearsch PA, Veloo ACM, Rodriguez-Palacios A. The genus _Alistipes_: Gut bacteria with emerging implications to inflammation,


cancer, and mental health. Front Immunol. 2020;11:906. Article  CAS  PubMed  PubMed Central  Google Scholar  * Du F, Huang RF, Lin D, Wang YY, Yang XH, Huang XY, et al. Resveratrol improves


liver steatosis and insulin resistance in non-alcoholic fatty liver disease in association with the gut microbiota. Front Microbiol. 2021;12:611323. Article  PubMed  PubMed Central  Google


Scholar  * Li W, Zhang K, Yang H. Pectin alleviates high fat (lard) diet-induced nonalcoholic fatty liver disease in mice: Possible role of short-chain fatty acids and gut microbiota


regulated by pectin. J Agric Food Chem. 2018;66:8015–25. Article  CAS  PubMed  Google Scholar  * Carpino G, Del Ben M, Pastori D, Carnevale R, Baratta F, Overi D, et al. Increased liver


localization of lipopolysaccharides in human and experimental NAFLD. Hepatology. 2020;72:470–85. Article  CAS  PubMed  Google Scholar  * Fusco R, Siracusa R, Genovese T, Cuzzocrea S, Di


Paola R. Focus on the role of NLRP3 inflammasome in diseases. Int J Mol Sci. 2020;21:4223. Article  CAS  PubMed  PubMed Central  Google Scholar  * Jeena K, Liju VB, Kuttan R. A preliminary


13-week oral toxicity study of ginger oil in male and female Wistar rats. Int J Toxicol. 2011;30:662–70. Article  CAS  PubMed  Google Scholar  * Reagan-Shaw S, Nihal M, Ahmad N. Dose


translation from animal to human studies revisited. FASEB J. 2008;22:659–61. Article  CAS  PubMed  Google Scholar  Download references ACKNOWLEDGEMENTS This study was financed by the


Ministry of Science and Technology (Taiwan) (109-2327-B-002-005, 109-2314-B-002-103-MY3, 109-2314-B-002-064-MY3, 110-2327-B-002-007, 111-2628-B-002-047, and 111-2327-B-002-008) and the


National Science and Technology Council, Taiwan (NSTC 112-2327-B-002-009, 113-2321-B-002-026, and 113-2321-B-002-022). The authors acknowledge the research collaboration and technical


support provided by the National Human Microbiome Core Facility, Taiwan (NSTC 112-2740-B-A49-002 and 113-2740-B-A49-003). AUTHOR INFORMATION Author notes * These authors contributed equally:


Suraphan Panyod, Wei-Kai Wu. AUTHORS AND AFFILIATIONS * Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC Suraphan Panyod, Ya-Chi Hsieh, Yea-Jing


Tseng, Sin-Yi Peng, Rou-An Chen, Huai-Syuan Huang & Lee-Yan Sheen * Center for Food and Biomolecules, National Taiwan University, Taipei, Taiwan, ROC Suraphan Panyod & Lee-Yan Sheen


* Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC Suraphan Panyod, Yi-Hsun Chen, Chun-Jen Liu & Ming-Shiang Wu * Department of


Medical Research, National Taiwan University Hospital, Taipei, Taiwan, ROC Wei-Kai Wu * Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, ROC Wei-Kai Wu, 


Chun-Jen Liu & Ming-Shiang Wu * Bachelor Program of Biotechnology and Food Nutrition, National Taiwan University, Taipei, Taiwan, ROC Wei-Kai Wu * Division of Gastroenterology, Perelman


School of Medicine, University of Pennsylvania, Philadelphia, PA, USA Ting-Chin David Shen * Department of Food Science, Rutgers University, New Brunswick, NJ, USA Chi-Tang Ho * National


Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan, ROC Hsiao-Li Chuang * Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan City,


Taiwan, ROC Chi-Chang Huang * National Center for Food Safety Education and Research, National Taiwan University, Taipei, Taiwan, ROC Lee-Yan Sheen Authors * Suraphan Panyod View author


publications You can also search for this author inPubMed Google Scholar * Wei-Kai Wu View author publications You can also search for this author inPubMed Google Scholar * Ya-Chi Hsieh View


author publications You can also search for this author inPubMed Google Scholar * Yea-Jing Tseng View author publications You can also search for this author inPubMed Google Scholar *


Sin-Yi Peng View author publications You can also search for this author inPubMed Google Scholar * Rou-An Chen View author publications You can also search for this author inPubMed Google


Scholar * Huai-Syuan Huang View author publications You can also search for this author inPubMed Google Scholar * Yi-Hsun Chen View author publications You can also search for this author


inPubMed Google Scholar * Ting-Chin David Shen View author publications You can also search for this author inPubMed Google Scholar * Chi-Tang Ho View author publications You can also search


for this author inPubMed Google Scholar * Chun-Jen Liu View author publications You can also search for this author inPubMed Google Scholar * Hsiao-Li Chuang View author publications You


can also search for this author inPubMed Google Scholar * Chi-Chang Huang View author publications You can also search for this author inPubMed Google Scholar * Ming-Shiang Wu View author


publications You can also search for this author inPubMed Google Scholar * Lee-Yan Sheen View author publications You can also search for this author inPubMed Google Scholar CONTRIBUTIONS SP


designed, instructed and assisted the experiment, performed the bioinformatics and statistical analysis, and drafted the manuscript; WKW instructed the experiments and reviewed the


manuscript; YCH, YJT, SYP performed the animal experiments; RAC and HSH assisted animal experiment; YHC assisted animal and LPS quantification experiment; TCDS, CTH, CJL, HLC, and CCH


critically reviewed the manuscript; MSW and LYS designed the experiments, provided the funding for the study, and critically revised the manuscript. CORRESPONDING AUTHORS Correspondence to


Ming-Shiang Wu or Lee-Yan Sheen. ETHICS DECLARATIONS COMPETING INTERESTS The authors declare no competing interests. ETHICS APPROVAL AND CONSENT TO PARTICIPATE Animal experiments were


approved by the Institutional Animal Care and Use Committee of National Taiwan University (IACUC) (Approval No: NTU-110-EL-00060). ADDITIONAL INFORMATION PUBLISHER’S NOTE Springer Nature


remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. SUPPLEMENTARY INFORMATION SUPPLEMENTARY FIGURE RIGHTS AND PERMISSIONS OPEN ACCESS This


article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction


in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the


licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article


are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and


your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this


licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/. Reprints and permissions ABOUT THIS ARTICLE CITE THIS ARTICLE Panyod, S., Wu, WK., Hsieh, YC. _et al._ Ginger essential oil


prevents NASH progression by blocking the NLRP3 inflammasome and remodeling the gut microbiota-LPS-TLR4 pathway in mice. _Nutr. Diabetes_ 14, 65 (2024).


https://doi.org/10.1038/s41387-024-00306-1 Download citation * Received: 09 August 2023 * Revised: 17 June 2024 * Accepted: 18 June 2024 * Published: 16 August 2024 * DOI:


https://doi.org/10.1038/s41387-024-00306-1 SHARE THIS ARTICLE Anyone you share the following link with will be able to read this content: Get shareable link Sorry, a shareable link is not


currently available for this article. Copy to clipboard Provided by the Springer Nature SharedIt content-sharing initiative