Open Access

Ophiopogonin D ameliorates non‑alcoholic fatty liver disease in high‑fat diet‑induced obese mice by improving lipid metabolism, oxidative stress and inflammatory response

  • Authors:
    • Xi Huang
    • Qi Ji
    • Chen-Yi She
    • Yi Cheng
    • Jian-Rong Zhou
    • Qing-Ming Wu
  • View Affiliations

  • Published online on: July 13, 2023     https://doi.org/10.3892/etm.2023.12116
  • Article Number: 418
  • Copyright: © Huang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lipid metabolic disorders, oxidative stress and inflammation in the liver are key steps in the progression of non‑alcoholic fatty liver disease (NAFLD). Ophiopogonin D (OP‑D), the main active ingredient of Ophiopogon japonicus, exhibits several pharmacological activities such as antioxidant and anti‑inflammatory activities. Therefore, the current study aimed to explore the role of OP‑D in NAFLD in a high‑fat diet (HFD)‑induced obesity mouse model. To investigate the effect of OP‑D on NAFLD in vivo, a NAFLD mouse model was established following feeding mice with HFD, then the mice were randomly treated with HFD or HFD + OP‑D for 4 weeks. Subsequently, primary mouse hepatocytes were isolated, and enzyme‑linked immunosorbent assay, reverse transcription‑quantitative PCR western blotting and immunofluorescence analysis were used for assessment to explore the direct effect of OP‑D in vitro. The results of the present study indicated that OP‑D could ameliorate NAFLD in HFD‑induced obese mice by regulating lipid metabolism and antioxidant and anti‑inflammatory responses. Additionally, OP‑D treatment decreased lipogenesis and inflammation levels in vitro, suggesting that the NF‑κB signaling pathway may be involved in the beneficial effects of OP‑D on NAFLD.

Introduction

Non-alcoholic fatty liver disease (NAFLD) represents a spectrum of liver injury diseases, including simple steatosis, non-alcoholic steatohepatitis as well as fibrosis and liver cirrhosis (1). Notably, the progressive form of NAFLD can lead to hepatocellular carcinoma and liver-related mortality (2,3). With the continuous increase in the prevalence of metabolic syndromes, such as type 2 diabetes mellitus (T2DM), obesity and hyperlipidemia, the morbidity of NAFLD has increased in parallel, affecting ~25% of the population worldwide, thus significantly contributing to the disease burden (2,4). At present, NAFLD is considered to be one of the most common chronic liver diseases worldwide (5). However, to date, there have been no approved pharmacological approaches for this condition. Therefore, the development of novel therapeutic strategies to improve the progression of NAFLD is important.

The pathogenesis and treatment of NAFLD have been widely investigated; however, they are still not entirely understood. The most accepted mechanism of NAFLD is the ‘multiple hits’ theory. According to this theory, NAFLD can be caused by several factors, such as insulin resistance (IR), dyslipidemia, oxidative stress (OS), endoplasmic reticulum stress and inflammation (6,7). Notably, dyslipidemia is a primary feature of NAFLD (8), while OS (free radical damage) and inflammation are considered as the most significant mechanisms leading to hepatic cell death and tissue injury (9,10). Therefore, regulating lipid metabolism, OS and the inflammatory response may be a particular approach for ameliorating the progression of NAFLD.

Ophiopogonin D (OP-D) is a significant pharmacological ingredient of the traditional Chinese medicine Ophiopogon japonicus. It has been reported that OP-D exhibits several pharmacological effects, including antioxidant and anti-inflammatory activities, while it also inhibits venous thrombosis (11-13). A recent study showed that OP-D can improve renal function by inhibiting OS and inflammation in streptozotocin-induced diabetic nephropathy rats (14). Therefore, the current study aimed to explore the role of OP-D on NAFLD in a mouse model of high-fat diet (HFD)-induced obesity.

Materials and methods

Drugs and reagents

OP-D was obtained from Shanghai Yuanye Biotechnology Co., Ltd. Palmitic acid (PA) was purchased from Sigma-Aldrich (Merck KGaA).

Animal experiments

A total of 15 male C57BL/6J mice (age, 3-4 weeks; weight, 17-20 g) were purchased from Shanghai Model Organisms Centre, Inc. Mice with similar body weights were allowed to adjust to the environment for one week prior to the experiments. Subsequently, mice (age, 4-5 weeks) were randomly divided into the following three groups: Normal chow diet (ND); HFD; and HFD + OP-D group. Mice were given free access to food and distilled water. Mice fed with ND (cat. no. D12450B; 10% fat, 70% carbohydrates and 20% protein; Research Diets, Inc.) or HFD (cat. no. D12492; 60% fat, 20% carbohydrate and 20% protein; Research Diets, Inc.). Following feeding for 12 weeks, HFD-induced obese mice were randomly allocated to the test groups for pharmacological studies. Mice in the HFD + OP-D group continued to be fed with HFD and were intragastrically administered with 5 mg/kg/day OP-D (dissolved in 0.5% of carboxymethyl cellulose) for the following 4 weeks, according to preliminary experiments and previous studies (14,15). Mice in the other groups were treated with the same volume of 0.5% sodium carboxyl methyl cellulose solution (in order to protect the gastric mucosa of mice) according to a previous study (16,17). At the end of the experiment (16 weeks), all mice were sacrificed by cervical dislocation. All mice were housed in specific pathogen-free facility under a 12-h light/dark cycle, a relative humidity of 50% and a controlled temperature of 22±1˚C. All animal procedures were approved by the Medical Ethics Committee of Wuhan University of Science and Technology (Wuhan, China; approval number, 2022139).

Serum biochemical measurements

At the end of the experiment, mice were fasted overnight and blood samples from the tail were collected to detect blood glucose levels using a blood glucose meter (Bayer AG). The mice were then sacrificed, and blood samples were collected by aortic puncture with centrifugation (1,300 x g for 10 min at 4˚C) to obtain serum, and stored at -80˚C for the blood chemistry measurements. The levels of insulin, IL-6, IL-1β and TNFα in serum were detected using the corresponding commercially available ELISA kits (insulin: cat. no. KCD-E1025; IL-6: cat. no. KCD-E1002; IL-1β: cat. no. KCD-E10010; TNFα: cat. no. KCD-E1018; Shanghai Kechuangda Biomedical Technology Co., Ltd.), according to the manufacturer's instructions. The serum levels of triglycerides (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were measured using an automatic biochemical analyzer (Hitachi, Ltd.). In addition, the serum levels of free fatty acids (FFA) were detected by a commercially available colorimetric assay kit (cat. No. A042-1-1, Nanjing Jiancheng Bioengineering Institute). The homeostatic model assessment for insulin resistance (HOMA-IR) index was calculated as follows: HOMA-IR=[fasting serum glucose (mmol/l) x fasting serum insulin (µU/ml)]/22.5.

Liver histology

Liver tissues from mice in each group were collected and fixed in 10% formaldehyde neutral buffer solution for 24 h at 4˚C, followed by washing with tap water, dehydration in alcohol and embedding in paraffin. Subsequently, the 5-µm thick transversal sections were obtained, deparaffinized, dehydrated in ethanol (50-100%) and cleared with xylene. The sections were stained using hematoxylin and eosin (H&E) solution for 5 min at room temperature (Wuhan Servicebio Technology Co., Ltd.), according to the manufacturer's protocols. For Oil-red O staining, liver tissues were frozen in optimal cutting temperature compound at -20˚C and 10-µm thick sections were obtained, followed by staining with Oil-red O solution (Wuhan Servicebio Technology Co., Ltd.) for 10 min at room temperature, according to the manufacturer's protocols. Images were observed using a NIKON imaging workstation (NIKON digital sight DS-FI2; Nikon Corporation).

Hepatic biochemical analysis

The commercially available Triglyceride Assay Kit (cat. no. ab65336; Abcam) was used to assess the levels of liver TG by measuring the optical density at a wavelength of 570 nm using spectrophotometry. The index of OS, including malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-Px) levels, was determined using the corresponding commercially available kits (MDA: cat. no. A003-1-1; SOD: cat. no. A001-1-1; CAT: cat. no. A007-1-1; GSH-Px: cat. no. A005-1-1. Nanjing Jiancheng Bioengineering Institute).

Cell cultures and treatments

Primary hepatocytes (PMHs) were isolated from mice. Briefly, 6-8-week-old male C57BL/6J mice (n=6; weight, 18-22 g) were purchased from Shanghai Model Organisms Centre, Inc. Mice were given free access to food and distilled water, and were housed in a specific pathogen-free facility under a 12-h light/dark cycle, a relative humidity of 50% and a controlled temperature of 22±1˚C. Mice were anesthetized using intraperitoneal injections of sodium pentobarbital (50 mg/kg). After the liver tissue was separated, the mice were sacrificed by cervical dislocation. The liver tissue was then collected, minced and filtered after portal vein perfusion. PMHs were purified and cultured in DMEM (cat. no. G4510; Wuhan Servicebio Technology Co., Ltd.) supplemented with 1% penicillin/streptomycin solution and 10% FBS (cat. no. G0004; Wuhan Servicebio Technology Co., Ltd.). To assess cell viability, PMHs (5x103) were seeded into 96-well plates for 24 h at 37˚C. The following day, when plates reached ~80% confluency, cells were treated with various concentrations (0-20 µmol/l) of OP-D for an additional 24 h at 37˚C. Cell viability was assessed using a Cell Counting Kit-8 assay (Wuhan Servicebio Technology Co., Ltd.). Briefly, 10 µl CCK-8 solution added into each well, which were then incubated at 37˚C with 5% CO2 for 2 h. The optical density was measured at a wavelength of 450 nm. To establish an in vitro model of lipid accumulation, hepatocytes were treated with PA (400 µmol/l) for 24 h at 37˚C. Then the cells were treated with or without 10 mol/l OP-D for another 24 h. Cultured cells were collected, isopropyl alcohol was added into 2x106 cells according to the proportion of 200 µl homogenizing medium, and centrifugation was performed at 4˚C and 10,000 x g for 10 min to obtain the lysed cells. The content of TG in lysed cells was measured using the TG Colorimetric Assay kit (cat. no. E-BC-K261-M; Elabscience Biotechnology, Inc.) following manufacturer's protocols. The optical density was measured at a wavelength of 510 nm.

Measurements of cytokine levels in culture supernatants

The levels of TNF-α, IL-1β and IL-6 in PMH supernatants were determined using ELISA, according to the manufacturer's instructions (Shanghai Kechuangda Biomedical Technology Co., Ltd.).

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from liver tissues or cells and isolated using TRIzol® reagent (Thermo Fisher Scientific, Inc.) according to the protocol provided by the manufacturer. The isolated RNA was reverse transcribed into cDNA using the RevertAid First Strand cDNA Synthesis Kit (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. qPCR was performed using SYBR Green PCR master mix (Applied Biosystems; Thermo Fisher Scientific, Inc.). The primer sequences used are listed in Table SI. All reactions were conducted in triplicate and the thermal cycling conditions were as follows: 30 sec at 95˚C, followed by 50 cycles of 95˚C for 5 sec and 60˚C for 30 sec. The mRNA expression levels of target genes were calculated using the 2-ΔΔCq method and normalized to those of GAPDH (18).

Western blot analysis

Liver tissues or cells were lysed with RIPA buffer containing protease inhibitors (Roche Applied Science). A BCA kit (Beyotime Institute of Biotechnology) was used to detect protein concentrations. The extracted proteins (20 µg of each protein) were loaded onto 10% SDS-polyacrylamide gel electrophoresis, and transferred to polyvinylidene fluoride membranes (MilliporeSigma) after electrophoresis. Blots were blocked with 5% bovine serum albumin (cat. no. GC305006; Wuhan Servicebio Technology Co., Ltd.) at 4˚C for 2 h and then incubated with primary antibodies against GAPDH (1:2,000; cat. no. 5174), P65 (1:1,000; cat. no. 8242), phosphorylated (p-)P65 (Ser468) (1:1,000; cat. no. 3039), IκBα (1:1,000; cat. no. 9242), p-IκBα (1:1,000; cat. no. 2859), TNFα (1:1,000; cat. no. 3707) (all CST Biological Reagents Co., Ltd.) at 4˚C overnight, The membranes were then incubated with HRP-conjugated anti-IgG secondary antibodies (cat. no. A0192; 1:2,000; Beyotime Institute of Biotechnology) at room temperature for 45 min. GAPDH was used as an internal reference. The intensity of protein bands was assessed using the ImageJ software (version 1.8.0; National Institutes of Health).

Immunofluorescence (IF) analysis

IF staining was carried out according to standard procedures (19). Briefly, cells were first fixed with 95% ethanol for 15 min at -20˚C. The cells were then incubated and blocked with blocking solution (5% BSA/PBS) for 30 min at room temperature, followed by being incubated overnight with a primary antibody against p65 (1:200; cat. no. 8242; Cell Signaling Technology, Inc.) at 4˚C. Subsequently, incubation with Alexa Fluor® 647-conjugated goat anti-rabbit IgG (1:400; cat. no. ab150079; Abcam) at 4˚C for 1 h was performed. The coverslips were mounted on microscope slides with fluorescence reagent containing 10 µg/ml DAPI (Thermo Fisher Scientific, Inc.). IF images were captured using the Fluoview FV1000 confocal microscope (Olympus Corporation).

Statistical data analysis

Data are presented as the mean ± SEM. All analyses were performed using GraphPad Prism 8.0 (GraphPad Software; Dotmatics) or SPSS 22.0 (IBM Corp.) software. Data among groups were compared using one-way ANOVA followed by Dunnett's test. P<0.05 was considered to indicate a statistically significant difference.

Results

OP-D attenuates body weight gain in HFD-treated mice

OP-D is a pharmacological compound of Ophiopogon japonicus, a traditional Chinese medicine herb (14). The molecular structure of OP-D is illustrated in Fig. 1A. The current study aimed to explore whether OP-D administration could inhibit body weight gain in mice fed with HFD. The results showed that the body weight of mice in the HFD + OP-D group was notably reduced compared with that in the HFD group at weeks 14-16 (Fig. 1B). Additionally, OP-D administration significantly reduced liver weight in HFD-treated mice compared with those in the HFD group (Fig. 1C).

OP-D improves glucose homeostasis and IR in mice fed with HFD

Subsequently, the present study investigated whether OP-D could regulate glucose and insulin homeostasis in HFD-fed mice. Consistent with previous studies (20,21), HFD-treated mice exhibited elevated glucose and insulin levels compared with ND control mice. The effect of the HFD on the levels of fasting blood glucose (FBG) and insulin was significantly reduced by OP-D treatment (Fig. 2A and B). Furthermore, the HOMA-IR index suggested that mice in the HFD + OP-D group displayed significantly reduced IR compared with those in the HFD group (Fig. 2C). These findings indicated that OP-D could exert a significant role in improving glucose homeostasis and IR in HFD-induced obese mice.

OP-D improves lipogenesis and hepatic steatosis in NAFLD mice

To further explore whether OP-D could prevent hepatic steatosis, H&E staining was first carried out. H&E staining showed widespread vacuolation in the liver tissue of HFD mice. However, OP-D treatment alleviated hepatic steatosis (Fig. 3A). In addition, mice in the HFD + OP-D group displayed a smaller area of hepatic lipid droplets compared with mice in the HFD group (Fig. 3A). Treatment with OP-D significantly reduced TG levels in the liver compared with the HFD group, thus suggesting that OP-D could attenuate lipid accumulation (Fig. 3B). Furthermore, TC, TG, FFA and LDL-C levels were significantly reduced in the serum of HFD mice treated with OP-D compared with mice treated with vehicle (Fig. 3C-F). Subsequently, the mRNA expression levels of lipid metabolism-related genes were detected. The data showed that the expression levels of lipid droplet formation-related genes [3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR), fat storage-inducing transmembrane protein (Fitm)1 and Fitm2], lipid uptake-related genes [fatty acid-binding protein 1 (Fabp1), fatty acid transporter 1 (Fatp1) and lipoprotein lipase (Lpl)] and lipogenesis-related genes [fatty acid synthase (FAS), stearoyl-CoA desaturase 1 (SCD1) and peroxisome proliferator-activated receptor (PPAR)-γ] were significantly decreased by treatment of HFD-fed mice with OP-D (Fig. 3G-I). By contrast, the expression of fatty acid β-oxidation-related genes [PPAR-α, peroxisomal acyl-coenzyme A oxidase 1 (Acox1) and carnitine O-palmitoyl transferase 1α (Cpt1α)] were significantly upregulated after OP-D treatment compared with the HFD group (Fig. 3J). Furthermore, the HFD-induced increased levels of ALT and AST in HFD mice were significantly reduced by OP-D (Fig. 3K and L). Taken together, these results suggested that OP-D could exert a protective role in the development of NAFLD.

OP-D attenuates OS and inflammation in NAFLD mice

Lipid peroxidation, OS and inflammation serve a critical role in the progression of NAFLD (22). Therefore, the levels of OS in the liver tissue were determined in the present study. The results revealed that the hepatic levels of MDA, an oxidative marker, were significantly reduced following treatment of HFD-induced NAFLD mice with OP-D (Fig. 4A). Inversely, the levels of the antioxidative markers SOD, CAT and GSH-Px were significantly increased in the livers of mice in the HFD + OP-D group compared with the HFD group that did not receive OP-D treatment (Fig. 4B-D). Furthermore, the mRNA expression levels of nuclear respiratory factor 1 and transcription factor A mitochondria, two mitochondrial regulators, were significantly increased in the HFD + OP-D group compared with the HFD group (Fig. 4E). Subsequently, the inflammatory response in mice treated with or without OP-D was investigated. As shown in Fig. 4F-H, the serum levels of the cytokines TNFα, IL-6 and IL-1β were significantly increased in HFD mice compared with ND mice. However, the secretion levels of these inflammatory mediators were restored following OP-D administration. Additionally, OP-D administration significantly reversed the increased mRNA expression levels of TNFα, IL-6 and Il-1β after induction by HFD (Fig. 4I-K). Furthermore, the significantly increased gene expression levels of the inflammatory mediators, including matrix metalloproteinase 9 (MMP9), monocyte chemoattractant protein 1 (MCP1) and vascular cell adhesion protein 1 (VCAM1), were also significantly reduced in the HFD + OP-D group compared with the HFD group (Fig. 4L). These findings suggested that OP-D could reduce OS and inflammation in HFD-induced NAFLD mice.

OP-D alleviates lipogenesis and inflammatory responses in vitro

The current study aimed to explore whether OP-D could exert a direct effect on PMHs. Therefore, the changes in fat deposition were evaluated in PMHs treated with different concentrations of PA for different time points (data not shown). Consistently, treatment with 400 µmol/l PA for 24 h was the optimum condition, as previously described (23). Since the CCK8 result showed that 0-20 µmol/l OP-D did not affect the cell viability of PMHs, the concentration of 10 µmol/l OP-D for 24 h was selected as the optimal condition (Fig. 5A). Treatment with OP-D significantly reduced TG levels in lysed PMHs compared with the PA-only treated group (Fig. 5B). The mRNA expression levels of lipid synthesis-related genes, such as acetyl-CoA carboxylase 1 (ACC1), FAS and SCD1, were significantly reduced in cells treated with OP-D compared with the PA group (Fig. 5C). Consistent with previous studies (11,24), PA significantly increased the levels of TNFα, IL-1β and IL-6 from PMHs, while OP-D reversed this effect (Fig. 5D-F). Additionally, OP-D could significantly reduce the mRNA expression levels of TNFα, IL-6 and IL-1β compared with the PA-only treated group (Fig. 5G). Collectively, these results indicated that OP-D could alleviate lipogenesis and inflammation in vitro.

NF-κB signaling is involved in the regulation of OP-D in PMHs

The results suggested that OP-D could alleviate inflammatory responses, which have a significant effect on the progression of HFD-induced NAFLD in mice. Therefore, the present study further explored the mechanisms underlying the effect of OP-D on NAFLD in PMHs. It has been reported previously that the inflammatory NF-κB signaling pathway plays a crucial role in regulating inflammatory responses in NAFLD (25). In the current study, western blot analysis demonstrated that cell treatment with OP-D significantly reduced the PA-induced protein expression levels of p-P65, p-IκBα and TNFα in hepatocytes (Fig. 6A-D). OP-D also inhibited the PA-induced nuclear translocation of P65 (Fig. 6E). These results suggested that the NF-κB signaling pathway could mediate the beneficial effects of OP-D on NAFLD.

Discussion

NAFLD is a common pathological disease of the liver, affecting ~25% of the global population, which increases the risk of hepatic and extrahepatic complications (2,4). Although several phytochemicals are currently used worldwide for the clinical treatment of NAFLD (26,27), the efficacy and safety of these drugs still remains unknown. Therefore, studies on the mechanisms underlying the development of NAFLD and potential protective therapeutic interventions are important.

Ophiopogon japonicus is a common herb in traditional Chinese medicine, that has been widely used for thousands of years due to its high nutritional and medicinal value (11-13). However, the mechanisms underlying the effects of Ophiopogon japonicus still require investigation. Emerging evidence has suggested that OP-D, a steroidal glycoside of Ophiopogon japonicus, exhibits several biological activities. Previous studies have demonstrated that OP-D can exert an anti-inflammatory effect on PM2.5-injured alveolar epithelial cells (28), protect human endothelial cells from OS injury (29) and alleviate diabetic myocardial injuries by reducing lipid accumulation and mitochondrial injury (15).

The current study aimed to investigate the possible therapeutic effects of OP-D on HFD-induced NAFLD. The main findings of the present study were as follows: i) OP-D alleviated obesity in a mouse model; ii) OP-D potentially played a beneficial role in NAFLD via the regulation of lipogenesis, OS injury and inflammation; and iii) the NF-κB signaling pathway was potentially involved in the regulation of OP-D in NAFLD.

NAFLD, currently known as ‘metabolic dysfunction-associated fatty liver disease’, is considered a hepatic manifestation of metabolic syndrome (30). The disease is closely associated with metabolic disorders, such as obesity, dyslipidemia, IR and T2DM (31). Obesity and being overweight are the most common causes of metabolic diseases and NAFLD (32). The present study showed that OP-D suppressed HFD-induced body weight gain in mice, which prompts further research on the effects of OP-D on NAFLD. It has been widely reported that IR and dyslipidemia are key events in the progression of NAFLD (32-34). In the current study, the results revealed that OP-D improved lipid metabolic disorders and IR in HFD-induced obese mice. These findings were consistent with those of a previous study that demonstrated that OP-D can improve hyperglycemia in diabetic rats (14).

OS plays an essential role in the occurrence and progression of NAFLD (9,35). OS refers to the imbalance of oxidants and antioxidants, which can lead to severe failure of cell function and eventually to cell death (36). Previous studies have demonstrated that the levels of the hepatic oxidation markers MDA and ROS are increased, while those of the antioxidative stress markers CAT, GSH-Px and SOD are decreased in NAFLD (36-38). The results of the current study showed that OP-D reduced OS, as evidenced by the reduced activity of MDA, and increased antioxidant activities, as evidenced by the increased SOD, CAT and GSH-Px activities.

The regulation of inflammatory responses has been recognized as a pivotal pathway for maintaining homeostasis and preventing the progression of NAFLD (9,39,40). It has been reported that NF-κB signaling plays a critical role in the regulation of inflammation (25). Once the NF-κB pathway is activated, pro-inflammatory cytokines, such as TNFα, IL-1β and IL-6, are widely expressed (41). The present study demonstrated that OP-D alleviated inflammatory responses in HFD-induced obese mice and verified that its anti-inflammatory effect was associated with the NF-κB pathway in vitro.

The current study indicated the effect of OP-D on alleviating hepatic steatosis via antioxidant and anti-inflammatory responses through the NF-κB signaling pathway; however, there are still certain limitations. Firstly, the particular upstream pathways involved in OP-D promoting P65 activation were not elucidated, and it could not be excluded that other mechanisms, such as mitochondrial OS, autophagy and ferroptosis could be involved in the aforementioned process. Secondly, the effects of OP-D on other cell types, such as Kupffer cells and sinusoidal endothelial cells in the liver, were not investigated. Finally, OP-D significantly reduced body weight, thus supporting the notion of a reduction in obesity; however, it cannot be excluded that muscle may also be reduced by OP-D administration. Therefore, further studies on the roles of OP-D on muscle volume or muscle strength are needed. Overall, further evidence is needed to clarify the mechanism underlying the effect of OP-D on improving NAFLD.

It has been reported that OP-D is safe for clinical use and the drug has little toxicity in animal subacute toxicity experiments (42,43). However, a recent study reported that OP-D can cause hemolysis in Kunming mice, but there is no further data to confirm this (44). In the present study, no specific side effects of OP-D on the HFD mice were observed. The side effects of OP-D still need to be investigated further.

In summary, the present study demonstrated that OP-D could alleviate NAFLD in HFD-induced obese mice by improving lipogenesis, inflammation and oxidative stress injury. In addition, NF-κB signaling could play an essential role in the beneficial effects of OP-D on NAFLD. Therefore, the above findings indicated that OP-D could be a potential therapeutic agent for NAFLD.

Supplementary Material

A list of reverse transcription-quantitative PCR primers used in this study.

Acknowledgements

Not applicable.

Funding

Funding: This study was supported by grants from the School of Medicine, Wuhan University of Science and Technology (grant no. OHIC2019G03).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

XH and QJ conducted the animal experiments. CS and YC performed the in vitro experiments. XH and YC wrote the manuscript. JZ and QW designed the study and conducted data analysis. QW is the guarantor of this work. XH and QW confirm the authenticity of all the raw data All authors have read and approved the final manuscript.

Ethics approval and consent to participate

All animal procedures were approved by the Medical Ethics Committee of Wuhan University of Science and Technology (Wuhan, China; approval no. 2022139).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Kumar S, Duan Q, Wu R, Harris EN and Su Q: Pathophysiological communication between hepatocytes and non-parenchymal cells in liver injury from NAFLD to liver fibrosis. Adv Drug Deliv Rev. 176(113869)2021.PubMed/NCBI View Article : Google Scholar

2 

Liebe R, Esposito I, Bock HH, Vom Dahl S, Stindt J, Baumann U, Luedde T and Keitel V: Diagnosis and management of secondary causes of steatohepatitis. J Hepatol. 74:1455–1471. 2021.PubMed/NCBI View Article : Google Scholar

3 

Safari Z and Gérard P: The links between the gut microbiome and non-alcoholic fatty liver disease (NAFLD). Cell Mol Life Sci. 76:1541–1558. 2019.PubMed/NCBI View Article : Google Scholar

4 

Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M, Harrison SA, Brunt EM and Sanyal AJ: The diagnosis and management of nonalcoholic fatty liver disease: Practice guidance from the American Association for the Study of Liver Diseases. Hepatology. 67:328–357. 2018.PubMed/NCBI View Article : Google Scholar

5 

Lan T, Hu Y, Hu F, Li H, Chen Y, Zhang J, Yu Y, Jiang S, Weng Q, Tian S, et al: Hepatocyte glutathione S-transferase mu 2 prevents non-alcoholic steatohepatitis by suppressing ASK1 signaling. J Hepatol. 76:407–419. 2022.PubMed/NCBI View Article : Google Scholar

6 

Rohm TV, Meier DT, Olefsky JM and Donath MY: Inflammation in obesity, diabetes, and related disorders. Immunity. 55:31–55. 2022.PubMed/NCBI View Article : Google Scholar

7 

Khan RS, Bril F, Cusi K and Newsome PN: Modulation of insulin resistance in nonalcoholic fatty liver disease. Hepatology. 70:711–724. 2019.PubMed/NCBI View Article : Google Scholar

8 

Katsiki N, Mikhailidis DP and Mantzoros CS: Non-alcoholic fatty liver disease and dyslipidemia: An update. Metabolism. 65:1109–1123. 2016.PubMed/NCBI View Article : Google Scholar

9 

Farzanegi P, Dana A, Ebrahimpoor Z, Asadi M and Azarbayjani MA: Mechanisms of beneficial effects of exercise training on non-alcoholic fatty liver disease (NAFLD): Roles of oxidative stress and inflammation. Eur J Sport Sci. 19:994–1003. 2019.PubMed/NCBI View Article : Google Scholar

10 

Mohammed S, Nicklas EH, Thadathil N, Selvarani R, Royce GH, Kinter M, Richardson A and Deepa SS: Role of necroptosis in chronic hepatic inflammation and fibrosis in a mouse model of increased oxidative stress. Free Radic Biol Med. 164:315–328. 2021.PubMed/NCBI View Article : Google Scholar

11 

Huang X, Wang Y, Zhang Z, Wang Y, Chen X, Wang Y and Gao Y: Ophiopogonin D and EETs ameliorate Ang II-induced inflammatory responses via activating PPARα in HUVECs. Biochem Biophys Res Commun. 490:123–133. 2017.PubMed/NCBI View Article : Google Scholar

12 

Kou J, Tian Y, Tang Y, Yan J and Yu B: Antithrombotic activities of aqueous extract from Radix Ophiopogon japonicus and its two constituents. Biol Pharm Bull. 29:1267–1270. 2006.PubMed/NCBI View Article : Google Scholar

13 

Kou J, Sun Y, Lin Y, Cheng Z, Zheng W, Yu B and Xu Q: Anti-inflammatory activities of aqueous extract from Radix Ophiopogon japonicus and its two constituents. Biol Pharm Bull. 28:1234–1238. 2005.PubMed/NCBI View Article : Google Scholar

14 

Qiao Y, Jiao H, Wang F and Niu H: Ophiopogonin D of Ophiopogon japonicus ameliorates renal function by suppressing oxidative stress and inflammatory response in streptozotocin-induced diabetic nephropathy rats. Braz J Med Biol Res. 53(e9628)2020.PubMed/NCBI View Article : Google Scholar

15 

Li W, Ji L, Tian J, Tang W, Shan X, Zhao P, Chen H, Zhang C, Xu M, Lu R and Guo W: Ophiopogonin D alleviates diabetic myocardial injuries by regulating mitochondrial dynamics. J Ethnopharmacol. 271(113853)2021.PubMed/NCBI View Article : Google Scholar

16 

Qin Y, Dong H, Sun J, Zhang Y, Li J, Zhang T, Chen G, Wang S, Song S, Wang W, et al: Evaluation of MTBH, a novel hesperetin derivative, on the activity of hepatic cytochrome P450 isoform in vitro and in vivo using a cocktail method by HPLC-MS/MS. Xenobiotica. 51:1389–1399. 2021.PubMed/NCBI View Article : Google Scholar

17 

Ma L, Wei S, Yang B, Ma W, Wu X, Ji H, Sui H and Chen J: Chrysosplenetin inhibits artemisinin efflux in P-gp-over-expressing Caco-2 cells and reverses P-gp/MDR1 mRNA up-regulated expression induced by artemisinin in mouse small intestine. Pharm Biol. 55:374–380. 2017.PubMed/NCBI View Article : Google Scholar

18 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

19 

Donaldson JG: Immunofluorescence staining. Curr Protoc Cell Biol. Chapter 4(Unit 4.3)2001.PubMed/NCBI View Article : Google Scholar

20 

Zandani G, Anavi-Cohen S, Yudelevich T, Nyska A, Dudai N, Madar Z and Gorelick J: Chiliadenus iphionoides Reduces body weight and improves parameters related to hepatic lipid and glucose metabolism in a high-fat-diet-induced mice model of NAFLD. Nutrients. 14(4552)2022.PubMed/NCBI View Article : Google Scholar

21 

Wu YK, Ren ZN, Zhu SL, Wu YZ, Wang G, Zhang H, Chen W, He Z, Ye XL and Zhai QX: Sulforaphane ameliorates non-alcoholic fatty liver disease in mice by promoting FGF21/FGFR1 signaling pathway. Acta Pharmacol Sin. 43:1473–1483. 2022.PubMed/NCBI View Article : Google Scholar

22 

Arroyave-Ospina JC, Wu Z, Geng Y and Moshage H: Role of oxidative stress in the pathogenesis of non-alcoholic fatty liver disease: Implications for prevention and therapy. Antioxidants (Basel). 10(174)2021.PubMed/NCBI View Article : Google Scholar

23 

Zhang X, Shang X, Jin S, Ma Z, Wang H, Ao N, Yang J and Du J: Vitamin D ameliorates high-fat-diet-induced hepatic injury via inhibiting pyroptosis and alters gut microbiota in rats. Arch Biochem Biophys. 705(108894)2021.PubMed/NCBI View Article : Google Scholar

24 

Wang Y, Huang X, Ma Z, Wang Y, Chen X and Gao Y: Ophiopogonin D alleviates cardiac hypertrophy in rat by upregulating CYP2J3 in vitro and suppressing inflammation in vivo. Biochem Biophys Res Commun. 503:1011–1019. 2018.PubMed/NCBI View Article : Google Scholar

25 

De Gregorio E, Colell A, Morales A and Marí M: Relevance of SIRT1-NF-κB Axis as therapeutic target to ameliorate inflammation in liver disease. Int J Mol Sci. 21(3858)2020.PubMed/NCBI View Article : Google Scholar

26 

Bansod S, Saifi MA and Godugu C: Molecular updates on berberine in liver diseases: Bench to bedside. Phytother Res. 35:5459–5476. 2021.PubMed/NCBI View Article : Google Scholar

27 

Bagherniya M, Nobili V, Blesso CN and Sahebkar A: Medicinal plants and bioactive natural compounds in the treatment of non-alcoholic fatty liver disease: A clinical review. Pharmacol Res. 130:213–240. 2018.PubMed/NCBI View Article : Google Scholar

28 

Wang Y, Li D, Song L and Ding H: Ophiopogonin D attenuates PM2.5-induced inflammation via suppressing the AMPK/NF-κB pathway in mouse pulmonary epithelial cells. Exp Ther Med. 20(139)2020.PubMed/NCBI View Article : Google Scholar

29 

Qian J, Jiang F, Wang B, Yu Y, Zhang X, Yin Z and Liu C: Ophiopogonin D prevents H2O2-induced injury in primary human umbilical vein endothelial cells. J Ethnopharmacol. 128:438–445. 2010.PubMed/NCBI View Article : Google Scholar

30 

Eslam M, Sanyal AJ and George J: International Consensus Panel. MAFLD: A consensus-driven proposed nomenclature for metabolic associated fatty liver disease. Gastroenterology. 158:1999–2014.e1. 2020.PubMed/NCBI View Article : Google Scholar

31 

Godoy-Matos AF, Silva Júnior WS and Valerio CM: NAFLD as a continuum: From obesity to metabolic syndrome and diabetes. Diabetol Metab Syndr. 12(60)2020.PubMed/NCBI View Article : Google Scholar

32 

Wasilewska N and Lebensztejn DM: Non-alcoholic fatty liver disease and lipotoxicity. Clin Exp Hepatol. 7:1–6. 2010.PubMed/NCBI View Article : Google Scholar

33 

Fujii H and Kawada N: Japan Study Group of Nafld Jsg-Nafld. The role of insulin resistance and diabetes in nonalcoholic fatty liver disease. Int J Mol Sci. 21(3863)2020.PubMed/NCBI View Article : Google Scholar

34 

Rhee EJ: Nonalcoholic fatty liver disease and diabetes: An epidemiological perspective. Endocrinol Metab (Seoul). 34:226–233. 2019.PubMed/NCBI View Article : Google Scholar

35 

Ma Y, Lee G, Heo SY and Roh YS: Oxidative stress is a key modulator in the development of nonalcoholic fatty liver disease. Antioxidants (Basel). 11(91)2021.PubMed/NCBI View Article : Google Scholar

36 

Li Q, Tan JX, He Y, Bai F, Li SW, Hou YW, Ji LS, Gao YT, Zhang X, Zhou ZH, et al: Atractylenolide III ameliorates non-alcoholic fatty liver disease by activating Hepatic Adiponectin Receptor 1-Mediated AMPK Pathway. Int J Biol Sci. 18:1594–1611. 2022.PubMed/NCBI View Article : Google Scholar

37 

Irie M, Sohda T, Anan A, Fukunaga A, Takata K, Tanaka T, Yokoyama K, Morihara D, Takeyama Y, Shakado S and Sakisaka S: Reduced glutathione suppresses oxidative stress in nonalcoholic fatty liver disease. Euroasian J Hepatogastroenterol. 6:13–18. 2016.PubMed/NCBI View Article : Google Scholar

38 

Polimeni L, Del Ben M, Baratta F, Perri L, Albanese F, Pastori D, Violi F and Angelico F: Oxidative stress: New insights on the association of non-alcoholic fatty liver disease and atherosclerosis. World J Hepatol. 7:1325–1336. 2015.PubMed/NCBI View Article : Google Scholar

39 

Yang Y, Lu Y, Han F, Chang Y, Li X, Han Z, Xue M, Cheng Y, Sun B and Chen L: Saxagliptin regulates M1/M2 macrophage polarization via CaMKKβ/AMPK pathway to attenuate NAFLD. Biochem Biophys Res Commun. 503:1618–1624. 2018.PubMed/NCBI View Article : Google Scholar

40 

Luo P, Qin C, Zhu L, Fang C, Zhang Y, Zhang H, Pei F, Tian S, Zhu XY, Gong J, et al: Ubiquitin-Specific Peptidase 10 (USP10) inhibits hepatic steatosis, insulin resistance, and inflammation through Sirt6. Hepatology. 68:1786–1803. 2018.PubMed/NCBI View Article : Google Scholar

41 

Castoldi A, Naffah De Souza C, Câmara NO and Moraes-Vieira PM: The macrophage switch in obesity development. Front Immunol. 6(637)2015.PubMed/NCBI View Article : Google Scholar

42 

Tang XL, Lin Y, Wang YG and Gao Y: Effects of ophiopogonin D on fatty acid metabolic enzymes in cardiomyocytes. Zhongguo Zhong Yao Za Zhi. 46:3672–3677. 2021.PubMed/NCBI View Article : Google Scholar : (In Chinese).

43 

Wu FM, Yang HY, Yang RS, Li M, Bao XH and Zhou J: Study on Quality Evaluation of Ophiopogonis Radix in Sichuan. Zhong Yao Cai. 39:1803–1808. 2016.PubMed/NCBI(In Chinese).

44 

Xu HH, Jiang ZH, Sun YT, Qiu LZ, Xu LL, Tang XL, Ma ZC and Gao Y: Differences in the hemolytic behavior of two isomers in ophiopogon japonicus in vitro and in vivo and their risk warnings. Oxid Med Cell Longev. 2020(8870656)2020.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

September-2023
Volume 26 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Huang X, Ji Q, She C, Cheng Y, Zhou J and Wu Q: Ophiopogonin D ameliorates non‑alcoholic fatty liver disease in high‑fat diet‑induced obese mice by improving lipid metabolism, oxidative stress and inflammatory response. Exp Ther Med 26: 418, 2023
APA
Huang, X., Ji, Q., She, C., Cheng, Y., Zhou, J., & Wu, Q. (2023). Ophiopogonin D ameliorates non‑alcoholic fatty liver disease in high‑fat diet‑induced obese mice by improving lipid metabolism, oxidative stress and inflammatory response. Experimental and Therapeutic Medicine, 26, 418. https://doi.org/10.3892/etm.2023.12116
MLA
Huang, X., Ji, Q., She, C., Cheng, Y., Zhou, J., Wu, Q."Ophiopogonin D ameliorates non‑alcoholic fatty liver disease in high‑fat diet‑induced obese mice by improving lipid metabolism, oxidative stress and inflammatory response". Experimental and Therapeutic Medicine 26.3 (2023): 418.
Chicago
Huang, X., Ji, Q., She, C., Cheng, Y., Zhou, J., Wu, Q."Ophiopogonin D ameliorates non‑alcoholic fatty liver disease in high‑fat diet‑induced obese mice by improving lipid metabolism, oxidative stress and inflammatory response". Experimental and Therapeutic Medicine 26, no. 3 (2023): 418. https://doi.org/10.3892/etm.2023.12116