1. Introduction
Obesity, defined by the World Health Organization as “abnormal or excessive fat accumulation that may impair health”, is a widespread metabolic disorder with serious implications for global public health [
1,
2]. Since the 1980s, the global prevalence of obesity has nearly tripled, and current trends suggest that by 2030, around 38% of adults worldwide will be classified as overweight, with an estimated 20% categorized as obese [
3]. The pathogenesis of obesity is multifactorial, involving a complex interplay between intrinsic genetic predispositions and extrinsic environmental factors such as dietary intake, energy homeostasis, family history, lifestyle patterns, and psychosocial influences [
4]. The primary physiological hallmark of obesity is the excessive accumulation of adipose tissue, which results from a prolonged imbalance between energy intake, metabolic processes, and energy expenditure. Chronic consumption of an HFD contributes to prolonged positive energy balance, leading to the conversion of excess energy into triglycerides and their subsequent storage in adipose tissue, thereby promoting the onset and progression of obesity [
5].
Alterations in the structure and composition of the gut microbiota represent one of the key factors associated with obesity. The composition of intestinal flora in obese individuals is significantly different from that in normal-weight individuals. Compared with normal-weight individuals, the ratio of Firmicutes/Bacteroidetes (F/B) at the phylum level of intestinal microflora in obese individuals is generally increased. The study of twins by Turnbaugh et al. demonstrated that obese individuals exhibit an increase in the relative abundance of Firmicutes and a decrease in the abundance of Bacteroidetes in the intestinal microbiota, resulting in a significantly higher F/B ratio than their lean counterparts [
6]. Substantial evidence indicates that modulating gut microbiota through dietary intervention to promote lipid metabolism and weight loss constitutes a promising and feasible strategy for improving metabolic health [
2]. Bioactive compounds, including dietary fiber [
7,
8], tea polyphenols [
9], and probiotic formulations [
10], can mitigate the onset and progression of obesity and its related complications by regulating the intestinal microbiota.
Dietary fiber, which is not fully digestible or utilizable by the human body, contributes negligible caloric value and therefore serves as an effective strategy for improving dietary quality and reducing overall energy intake. Based on solubility, dietary fiber is categorized into SDF and insoluble dietary fiber (IDF). Compared with IDF, SDF is more readily fermented by intestinal microorganisms. It interacts with water and undergoes rapid fermentation and softening in the colon under microbial action, leading to the production of metabolites such as SCFAs, which exert beneficial physiological effects [
11]. These SCFAs inhibit hepatic lipid synthesis by activating the AMPK signaling pathway, inhibiting sterol regulatory element binding protein 1c (SREBP-1c) and 3-hydroxy-3-methyl glutaryl coenzyme A reductase (HMGCR) expression, reducing fatty acid synthase (FASN) and acetyl CoA carboxylase (ACC) activity [
12,
13]. By activating peroxisome proliferator-activated receptor alpha (PPARα), upregulating carnitine palmitoyltransferase 1 (CPT1), and enhancing mitochondrial fatty acid β-oxidation, SCFAs promote lipid oxidative breakdown [
14]. SCFAs also have the function of regulating adipose tissue by stimulating adipocytes to secrete leptin, inhibiting appetite and increasing energy consumption [
15]. In addition, SCFAs enter the blood through the colonic epithelium and affect lipid, glucose and cholesterol metabolism via interactions with G protein-coupled receptors [
16].
Highland barley (Qingke) is a unique cereal grain native to the Qinghai–Tibet Plateau region of China. Its bran, a major byproduct of highland barley flour processing, is rich in dietary fiber, with a content ranging from 58.7 to 65.2% dietary fiber-significantly higher than that of wheat bran (42.3%) [
17]. However, the dense structure of natural bran fiber results in a low SDF proportion of only 6.1–8.3%, limiting the full release of its functional activity. In our previous work, to enhance SDF extraction yield and improve its physical, structural, and functional properties, we developed a combined approach involving solid-state fermentation using
Lactobacillus bulgaricus and
Streptococcus thermophilus, followed by enzymatic extraction. This treatment significantly improved the functional characteristics of SDF, including enhanced water- and oil-holding capacity, increased solubility, a more porous and loose surface morphology, enlarged specific surface area, and improved abilities to adsorb cholesterol and bile salts. Moreover, the modified SDF exhibited notable lipid-lowering effects in vitro [
18]. Nevertheless, whether highland barley bran SDF exerts lipid-lowering effects in vivo and the underlying mechanisms remain unclear. To comprehensively investigate the in vivo hypolipidemic effects and mechanistic basis of SDF derived from highland barley bran, this study employs an HFD-induced obese mouse model, integrated with metabolomics, high-throughput sequencing, and molecular biology techniques. The findings aim to provide a scientific foundation and theoretical support for the development of functional products derived from highland barley bran SDF for obesity management.
2. Materials and Methods
2.1. Materials and Chemicals
The preparation of highland barley bran SDF followed the methodology described by Wei et al. [
18], utilizing a mixed fermentation process with
Lactobacillus bulgaricus and
Streptococcus thermophilus at a ratio of 4:2 (
w/
w), followed by enzymatic extraction to obtain SDF with a purity of 89%. The detailed process of preparing SDF was shown in the
Supplementary Figure S1.
2.2. Experimental Instruments
Scientific Multiskan Sky Automated Microplate Reader, Beijing Pengkun Boyuan Science and Technology Development Co., Ltd., Beijing, China; Tissuelyser-96 Tissue Homogenizer, Shanghai Jingxin Experimental Technology Co., Ltd., Shanghai, China; Analytical Balance, Langfang Zhongyi Technology Co., Ltd. Langfang, China; Eclipse Ci-L Upright Microscope-Nikon Corporation, Tokyo, Japan; SCIEX Triple Quad™ 6500+ Mass Spectrometer, SCIEX Corporation, Marlborough, MA, USA; Centrifuge, Heraeus Fresco 17, Thermo Fisher Scientific Inc., Waltham, MA, USA; CFX96TM Optics Module, CFX96TM Real-Time System, BIO-RAD Laboratories, Inc., Hercules, CA, USA; Electrophoresis System-Beijing Liuyi Biotechnology Co., Ltd., Beijing, China; Benchtop Vortex Mixer, AHN myLab® VT-03, AHN Biotechnologie GmbH, Nordhausen, Germany; Microplate Reader, Multiskan FC, Thermo Fisher Scientific Inc., Waltham, MA, USA.
2.3. Animals and Treatments
A total of eighteen male SPF-grade C57BL/6J mice, aged five weeks and weighing approximately 15 g, were obtained from Sibefu Biotechnology Co., Ltd. (Beijing, China) (Certification No. SCXK(Jing)2024-0001). The mice were maintained in a clean-grade barrier environment at the Qinghai Institute for Endemic Disease Prevention and Control, under standardized conditions including a temperature of 25 ± 2 °C, relative humidity of 50 ± 15%, and a 12 h light-dark cycle. Following a one-week acclimatization period, the mice were randomly assigned to three experimental groups (
n = 6 per group): the normal diet control group (ND group), the HFD group, and the SDF intervention group (SDF group), which received HFD supplemented with 0.5 g/kg body weight of SDF. The ND group received a purified diet with 10% fat-derived energy (
Supplementary Table S1) and regular drinking water, while the HFD and SDF groups were fed a high-fat purified diet providing 60% fat-derived energy (
Supplementary Table S1) along with regular drinking water. Following an 8-week feeding period, the mice were fasted overnight, anesthetized, and euthanized via cervical dislocation after blood collection from the orbital sinus. Blood samples were centrifuged at 3500 rpm for 15 min at 4 °C to collect serum. Liver tissues were carefully excised, washed with cold physiological saline to eliminate residual blood, and weighed accurately. Serum, liver specimens, and fecal samples were immediately frozen in liquid nitrogen and kept at −80 °C until further analysis.
2.4. Determination of Body Weight and Food Efficiency Ratio
The body weight and food intake of mice were recorded weekly and the food efficiency ratio was calculated according to Formula (1).
2.5. Assessment of Blood Lipid Levels
Assay kits supplied by Nanjing Jiancheng Bioengineering Institute (Nanjing, China) were utilized to determine the concentrations of total cholesterol (TC), total triglycerides (TG), high-density lipoprotein cholesterol (HDL-c), low-density lipoprotein cholesterol (LDL-c), alanine aminotransferase (ALT), and aspartate aminotransferase (AST) in mouse serum, with all procedures strictly following the manufacturer’s recommended guidelines.
2.6. Histological Examination of Liver Tissue
Mouse liver tissues were immersed in 4% paraformaldehyde at ambient temperature for 48 h, followed by dehydration via gradient ethanol solutions, xylene-mediated clearing for transparency, and subsequent embedding in paraffin wax. A microtome was employed to cut the paraffin-embedded specimens into 4-μm-thick sections, which were then subjected to hematoxylin and eosin (H&E) staining for histological examination, in accordance with the protocol described in Reference [
19].
2.7. The Liver Index of Mice
The hepatic index of mice was calculated according to Formula (2).
2.8. Determination of Hepatic Tissue Lipid Content
A 50 mg portion of liver tissue was weighed and homogenized in ice-cold physiological saline, then centrifuged at 1000×
g for 10 min. The supernatant was harvested and used to measure TG and TC concentrations according to the protocols provided with a commercial kit from Nanjing Jiancheng Bioengineering Institute (Nanjing, China) [
20]. Meanwhile, 50 mg of liver tissue was homogenized in absolute ethanol and centrifuged at 1000×
g for 10 min. The resulting supernatant was analyzed for AST and ALT concentrations using an assay kit from Nanjing Jiancheng Bioengineering Institute (Nanjing, China) [
21].
2.9. Hepatic Lipidomics
20 mg of liver tissue was weighed on dry ice and transferred to a tube containing steel beads. Subsequently, 400 μL of water was added, and the mixture was vortexed for 60 s, followed by homogenization at 45 Hz for 4 min and ultrasonic disruption for 5 min in an ice-water bath. This procedure was repeated three times. From the resulting homogenate, 50 μL was aliquoted into a new tube, combined with 150 μL of water, and then mixed with 800 μL of extraction solvent (MTBE: MeOH = 5:1, containing internal standards). The sample was vortexed for 60 s and then sonicated for 10 min in an ice-water bath. Following this, it was centrifuged at 3000 rpm (4 °C) for 15 min, and 500 μL of the resulting supernatant was collected. The supernatant was vacuum-dried at 37 °C, and the residue was reconstituted in 150 μL of reconstitution solution (DCM: MeOH: H
2O = 60:30:4.5). The mixture was vortexed for 30 s and ultrasonicated for 10 min in an ice-water bath. Following this, samples were centrifuged again at 12,000 rpm (4 °C) for 15 min. Finally, 70 μL of the resulting supernatant was transferred to an autosampler vial for LC-MS analysis. Furthermore, a quality control (QC) sample was created by pooling 20 μL of supernatant from each individual sample, and this pooled QC was analyzed concurrently with the experimental samples to monitor analytical consistency [
22].
Chromatographic separation of the target compounds was carried out using an ACQUITY Premier ultra-high performance liquid chromatography (UHPLC) system (ACQUITY Premier, Waters, Milford, CT, USA) [
23]. The mobile phase and flow rate were described in
Supplementary Table S2. Detection and method development were performed using a SCIEX Triple Quad™ 6500+ mass spectrometer equipped with an electrospray ionization source. Key source parameters included an IonSpray voltage of +5500 V (positive mode) or −4500 V (negative mode), curtain gas set at 35 psi, ion source gases 1 and 2 both at 50 psi, source temperature of 350 °C, and a declustering potential of ±80 V. Data acquisition and quantification were conducted using SCIEX Analyst Software (v1.6.3) in conjunction with DATA DRIVEN FLOW (v2.0.3.11). The absolute amount of each target lipid was determined by comparing its peak area to that of the corresponding class-specific internal standard.
2.10. RT-qPCR
Total RNA was extracted from mouse liver tissues using a total RNA extraction kit (Service Biotechnology Co., Ltd., Wuhan, China). The total RNA was reverse transcribed using a reverse transcription kit. The mRNA expression levels in mouse liver tissues were then detected by a two-step method. The PCR program was 30 s of pre-denaturation at 95 °C, 15 s of denaturation at 95 °C, and 30 s of annealing/extension at 60 °C for 40 cycles. The relative mRNA expression of each target gene was calculated using the 2
−ΔΔCt method, with primer sequences provided in
Supplementary Table S3.
2.11. Protein Extraction from Liver Tissues and Western Blot Assay
Liver tissue proteins were isolated and subjected to Western blot analysis in accordance with the protocol described in our previous research [
19]. Detailed information regarding the primary antibodies used in the present study is provided in
Supplementary Table S4.
2.12. Analysis of Gut Microbiota
One week prior to sacrifice, each mouse was individually housed in a sterile cage on a daily basis to allow for unrestricted fecal excretion. Fresh fecal samples were collected using sterile forceps and immediately transferred into 1.5 mL cryovials. Following the collection of 200 mg fresh fecal samples, total genomic DNA was extracted with a commercial DNA isolation kit (Service Biotechnology Co., Ltd., Wuhan, China), strictly in line with the manufacturer’s recommended procedures. The integrity of the extracted DNA was assessed via 1% agarose gel electrophoresis, while its purity was assessed using a microplate reader. Samples displaying distinct electrophoretic bands and intact genomic profiles, fulfilling the criteria for subsequent sequencing, had their concentrations quantified with a NanoDrop spectrophotometer. The V3–V4 hypervariable regions of the bacterial 16S rDNA were amplified via PCR using the primer pair 341F (CCTACGGGNGGCWGCAG) and 805R (GACTACHVGGGTATCTAATCC). The resulting PCR amplicons were verified by agarose gel electrophoresis and subsequently purified with the Qiagen Gel Extraction Kit (75510-019, Invitrogen, Carlsbad, CA, USA). High-throughput sequencing was carried out on the Illumina NovaSeq 6000 platform at Shanghai Baiqu Biomedical Technology Co., Ltd. (Shanghai, China), Raw sequence data were processed and analyzed using the bioinformatics pipeline detailed in our earlier studies [
24].
2.13. Determination of SCFAs in the Fecal Samples of Mice
Fecal samples were placed into 2 mL EP tubes and resuspended in 1 mL of ultrapure water, followed by brief vortex mixing (10 s). Steel beads were introduced, and the samples were mechanically lysed at 40 Hz for 4 min. After lysis, the suspensions underwent three cycles of ultrasonication on ice (4 °C, 5 min per cycle). The homogenates were then centrifuged at 5000 rpm and 4 °C for 20 min. A 0.8 mL aliquot of the supernatant from each sample was transferred into a new 2 mL tube. Thereafter, 0.1 mL of 50% sulfuric acid (H2SO4) and 0.8 mL of extraction solvent (methyl tert-butyl ether supplemented with 25 mg/L 2-methylvaleric acid as the internal standard) were added sequentially. The resultant mixture was vortexed for 10 s, oscillated for 10 min, and subjected to additional sonication on ice (4 °C, 10 min). Following incubation at −20 °C for 30 min to facilitate phase separation, the samples were centrifuged again at 10,000 rpm and 4 °C for 15 min. The upper organic layer was collected and transferred to a GC-MS vial, which was then prepared for subsequent detection and analysis.
A SHIMADZU GC2030-QP2020 NX system fitted with a capillary column (HP-FFAP, Agilent Technologies, Inc., Palo Alto, CA, USA) was employed for GC-MS analysis. The instrument parameter settings were shown in
Supplementary Table S5.
2.14. FMT
A total of eighteen male SPF-grade C57BL/6J donor mice, aged five weeks, were obtained from Sibefu Biotechnology Co., Ltd. (Beijing, China). The mice were maintained in a clean-grade barrier environment at the Qinghai Institute for Endemic Disease Prevention and Control, under standardized conditions including a temperature of 25 ± 2 °C, relative humidity of 50 ± 15%, and a 12 h light-dark cycle. Following a one-week acclimatization period, the mice were randomly assigned to three groups (n = 6 per group): ND, HFD and SDF group (received HFD supplemented with 0.5 g/kg body weight of SDF). The ND group received a purified diet with 10% fat-derived energy and regular drinking water, while the HFD and SDF groups were fed a high-fat purified diet providing 60% fat-derived energy along with regular drinking water. After four weeks of feeding, daily collection of fresh fecal samples was conducted from the ND, HFD, and SDF groups. 200 mg of fresh feces were suspended in 2 mL sterile saline solution. The solution was vigorously vortexed for 10 s using a benchtop vortex mixer, followed by centrifugation at 800× g for 3 min. The supernatant was collected as the transplantation material. To minimize changes in microbial composition, the fecal transplant suspension was freshly prepared within 10 min prior to oral administration.
A total of eighteen male SPF-grade C57BL/6J recipient mice, aged five weeks, were obtained from Sibefu (Beijing) Biotechnology Co., Ltd. (Beijing, China), and were maintained in a clean-grade barrier environment at the Qinghai Institute for Endemic Disease Prevention and Control, under standardized conditions including a temperature of 25 ± 2 °C, relative humidity of 50 ± 15%, and a 12 h light-dark cycle. After a one-week acclimatization period, the mice were administered a 200 μL cocktail of antibiotics (ampicillin 1 g/L, metronidazole 1 g/L, vancomycin 0.5 g/L, and neomycin 0.5 g/L) daily for two weeks to deplete the native gut microbiota. Following antibiotic pretreatment, the mice were randomly assigned to three groups. They were the FND (ND group-derived fecal microbiota), FHFD (HFD group-derived fecal microbiota), and FSDF (SDF group-derived fecal microbiota) groups, respectively. The recipient mice were fed a high-fat purified diet providing 60% fat-derived energy along with regular drinking water. Each group (
n = 6) received daily gavages of 100 μL of freshly prepared fecal suspension from donor mice of the ND, HFD, or SDF groups, respectively [
25,
26]. To reduce the differences among the donor microbial communities, equal amounts of feces from each donor mouse were collected and mixed together to prepare a bacterial suspension for FMT. The transplanted mice were housed in groups of four per cage. The study concluded after four weeks, at which point serum, liver tissue, fecal samples, and other specimens were collected and stored at −80 °C for further analysis.
2.15. Statistical Analyses
Data are expressed as mean ± standard deviation. Statistical analysis was performed using SPSS 26 software (IBM, Armonk, NY, USA), with the Tukey–Kramer method applied to determine significant differences, where p ≤ 0.05 was deemed statistically significant. Data analysis was performed using GraphPad Prism version 10.1.2 software (GraphPad, San Diego, CA, USA).
4. Discussion
This study demonstrates that soluble dietary fiber (SDF) ameliorates HFD-induced hepatic lipid metabolic disorders by activating the SCFA–pAMPK signaling pathway, thereby significantly reducing serum and hepatic lipid levels and improving liver function.
Liver lipidomics analysis was performed to investigate alterations in hepatic lipid metabolites in HFD-fed mice following intervention with SDF derived from highland barley bran. Three potential lipid biomarkers responsive to SDF treatment were identified: PE (18:2–20:3), PS (18:0–18:2), and PS (18:1–22:3). PE is closely associated with liver cell regeneration. Carril et al. reported that an increased hepatic PE content promoted liver tissue regeneration in a mouse model of non-alcoholic steatohepatitis [
28]. In this study, HFD feeding led to significant lipid accumulation, extensive lipid droplet deposition, and hepatocellular injury. Following SDF intervention, the number of lipid droplets in mouse liver tissue was decreased, and hepatocytes exhibited morphological recovery toward normal status, which may be linked to an elevated proportion of PE in the liver tissue. PS has been shown to act as a PPARα agonist, activating PPARα signaling and promoting fatty acid oxidation [
29]. Therefore, SDF from highland barley bran increased the hepatic levels of PS (18:0–18:2) and PS (18:1–22:3) in HFD-fed mice, which was accompanied by upregulated PPARα expression and activation of CPT1, leading to enhanced lipid metabolism.
A well-balanced gut microbial community is essential for the prevention and management of obesity. The mechanisms by which gut microbiota modulation ameliorates obesity include maintaining intestinal barrier integrity and regulating key microbial metabolites, particularly SCFAs. SCFAs can enter the systemic circulation and directly or indirectly activate AMPK in hepatocytes [
30,
31]. For instance, propionic acid activates the AMPK signaling pathway, suppresses the expression of key lipogenic regulators such as HMGCR and SREBP-1c, and reduces the enzymatic activities of FASN and ACC, thereby inhibiting hepatic lipid synthesis [
12,
13]. Meanwhile, butyrate activates PPARα, leading to the upregulation of CPT1, enhanced mitochondrial fatty acid β-oxidation, and increased lipid oxidative breakdown [
14]. Thus, the gut microbiota can indirectly regulate hepatic lipid metabolism through the SCFA-AMPK signaling axis. In this study, supplementation with SDF from highland barley bran significantly increased the abundance of beneficial bacteria, including Actinobacteriota,
Dubosiella,
Lactobacillus, and
Bifidobacterium. Actinobacteriota have been reported to exert beneficial effects on host health [
32].
Bifidobacterium is a key probiotic genus within the phylum Actinobacteria. Previous studies have shown that dietary supplementation with
Bifidobacterium in HFD-fed C57BL/6J mice improves glucose tolerance and insulin sensitivity, and enhances acetic acid production [
33]. These SCFAs are essential intestinal metabolites that confer beneficial effects on host health, including reduction of body weight in obese mice, amelioration of glucose and lipid metabolism disorders, and alleviation of hepatic steatosis [
32]. Consistent with prior findings—such as those showing hazelnut SDF ameliorates hyperlipidemia and obesity in rats by enriching
Lactobacillus and
Roseburia [
34], and bamboo shoot dietary fiber increasing
Bifidobacterium and
Dubosiella abundance in obese mice [
32]—our results demonstrate that highland barley bran SDF significantly elevates fecal concentrations of total SCFAs and acetic acid in HFD-fed mice. Notably, SDF intervention significantly upregulated hepatic protein expression levels of pAMPK and total AMPK (
p < 0.05), indicating enhanced activation of pAMPK in the liver. Furthermore, SDF downregulated the protein expression of key lipogenic genes, including HMGCR, SREBP-1c, FAS, ACC, and SCD1. Concurrently, SDF upregulated the expression of CPT1α and PPARα—critical regulators of fatty acid transport into mitochondria and subsequent β-oxidation—thereby promoting hepatic lipid catabolism [
35]. Supporting evidence from Zhou et al. also indicates that bamboo shoot dietary fiber alleviates obesity by modulating host PPAR signaling and fatty acid metabolism pathways [
32]. Based on these findings, we propose that SDF alleviates HFD-induced lipid metabolism disorders by enriching beneficial gut bacteria, boosting SCFA production, and consequently activating the hepatic AMPK signaling pathway.
To further confirm the mediating role of the gut microbiota in the ability of SDF from highland barley bran to ameliorate hepatic lipid metabolism disorders in HFD-fed mice, we conducted an FMT experiment. The results indicated that FMT effectively reshaped the structure and composition of the gut microbiota in recipient mice, with the microbial profile being similar to that of the donor group.
At the genus level, the relative abundance of Dubosiella was higher in the FSDF group compared to the FHFD group (p < 0.05), consistent with the elevated Dubosiella levels observed in the original SDF donor group relative to the HFD control group. Notably, the concentration of total SCFAs in the fecal samples of mice in the FSDF group was significantly increased. These findings collectively confirm that SDF improves hepatic lipid metabolism in HFD-fed mice by reshaping HFD-induced gut microbiota dysbiosis, elevating the relative abundance of beneficial gut bacteria (notably Dubosiella), thereby promoting SCFA biosynthesis and activating the SCFAs-AMPK signaling pathway. This study provides direct evidence for the causal role of gut microbiota in mediating the metabolic benefits of highland barley bran SDF.
This study still has many limitations. SCFAs have been established as important regulatory factors that act as signaling molecules via G-protein-coupled receptors, including free fatty acid receptor 2 (FFAR2) and FFAR3. However, this study did not investigate the impact of SDF on FFAR2 and FFAR3. In future studies, we will explore whether SCFA-mediated receptor signaling promotes AMPK activation and lipid metabolism. Additionally, although this study verified the mediating role of gut microbiota in the intervention of obesity by SDF through FMT, and clarified that highland barley bran SDF improved lipid metabolism disorders caused by an HFD by reshaping gut microbiota and activating the SCFAs-AMPK pathway. However, due to species differences, dietary doses, and the complexity of intestinal and liver signal transduction, the lipid-lowering mechanism of SDF needs to be further verified in clinical and cell experiments.