Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (298)

Search Parameters:
Keywords = fecal lipids

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
25 pages, 4126 KB  
Article
Structural Characterization of Polysaccharide from Flammulina velutipes and Its Impact on Hyperlipidemia Through Modulation of Hepatic Cholesterol Metabolism and Gut Microbiota
by Wei Jia, Huimin Wang, Ting Feng, Xiaoxiao Liu, Zhendong Liu, Zhengpeng Li, Wenhan Wang and Jingsong Zhang
Foods 2025, 14(19), 3452; https://doi.org/10.3390/foods14193452 - 9 Oct 2025
Viewed by 110
Abstract
FVPB1, a novel heteropolysaccharide, was extracted from the Flammulina velutipes fruiting body, and its structure was determined by methylation analysis, nuclear magnetic resonance (NMR) spectroscopy. FVPB1 demonstrated efficacy in inhibiting lipid accumulation in Raw264.7 cells and zebrafish, as well as in reducing weight [...] Read more.
FVPB1, a novel heteropolysaccharide, was extracted from the Flammulina velutipes fruiting body, and its structure was determined by methylation analysis, nuclear magnetic resonance (NMR) spectroscopy. FVPB1 demonstrated efficacy in inhibiting lipid accumulation in Raw264.7 cells and zebrafish, as well as in reducing weight gain and ameliorating liver injury in high-fat diet-induced mice. High concentration of FVPB1 significantly increased serum ApoA1 levels, while all tested doses (low, medium, and high) reduced serum ApoB levels in mice. Intervention with FVPB1 significantly increased the mRNA expression of Lcat and Cyp7a1 enzymes while markedly reducing the transcriptional level of Hmgcr reductase. Additionally, low concentration of FVPB1 enhanced CYP7A1 protein expression, whereas medium and high concentrations of FVPB1 promoted LCAT protein expression. Medium and high concentrations of FVPB1 significantly enhanced bile acid excretion in mice, with the high dose additionally promoting fecal sterol output. Alpha and beta diversity analyses demonstrated that a high-fat diet induced substantial dysbiosis in the gut microbiota of mice, characterized by reduced microbial diversity and richness. Intervention with FVPB1 significantly modulated the structural composition of the intestinal microbiota in high-fat diet-fed mice. Therefore, FVPB1 exerts lipid-lowering effect in high-fat diet-fed mice by modulating cholesterol metabolism and ameliorating gut microbiota dysbiosis. Full article
(This article belongs to the Section Nutraceuticals, Functional Foods, and Novel Foods)
Show Figures

Figure 1

14 pages, 2769 KB  
Article
Dietary Mannan Oligosaccharides Enhance Lactational Performance, Nutrient Metabolism, Plasma Metabolomics, and Gut Microbiota in Dezhou Donkeys
by Tianzheng Wang, Yaru Wang, Pengshuai Li, Jiaxin Liu, Xinyi Mao, Zuowei Li, Zhangxinhan Wen, Yuhan Yin, Yan Li, Gang Lin, Haihua Zhang, Honglei Qu, Qiugang Ma and Shimeng Huang
Int. J. Mol. Sci. 2025, 26(18), 9105; https://doi.org/10.3390/ijms26189105 - 18 Sep 2025
Viewed by 362
Abstract
This study investigated the effects of dietary mannan oligosaccharide (MOS) supplementation on growth performance, serum biochemistry, metabolomic profiles, and fecal microbiota in lactating Dezhou donkeys. Sixteen healthy jennies and their foals were randomly allocated to a control group (MCON), a group receiving no [...] Read more.
This study investigated the effects of dietary mannan oligosaccharide (MOS) supplementation on growth performance, serum biochemistry, metabolomic profiles, and fecal microbiota in lactating Dezhou donkeys. Sixteen healthy jennies and their foals were randomly allocated to a control group (MCON), a group receiving no MOS, or an MOS-supplemented group (MMO; 0.5 g/kg diet) for 60 days. Compared with the MCON group, the MMO group showed a mitigation of lactational weight reduction, improved serum protein profiles, and favorable modulation of lipid metabolism. Furthermore, serum metabolomic analysis revealed 102 differentially abundant metabolites, which were enriched in 17 KEGG pathways involved in energy metabolism, bile secretion, and anti-inflammatory signaling. Key metabolites such as L-4-Chlorotryptophan, Gly-Trp, and cholylthreonine indicated enhanced nutrient metabolism and gut barrier function. Moreover, MOS supplementation significantly increased alpha diversity of the gut microbiota, altered community composition, and promoted the abundance of beneficial genera, including Clostridium and Bacteroides. Collectively, these results demonstrate that MOS supplementation improves metabolic health, modulates immune and antioxidant responses, and fosters a beneficial gut microbial ecosystem in lactating donkeys, suggesting its potential as an effective prebiotic in equine nutrition. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

17 pages, 2183 KB  
Article
Anti-Obesity Effects and Changes of Fecal Microbiome by Lactic Acid Bacteria from Grains in a High-Fat Diet Mouse Model
by Chang Woo Jeon, Hyeon Yeong Lee, Hong Sik Kim, Min Ju Seo, Kye Won Park and Jung-Hoon Yoon
Int. J. Mol. Sci. 2025, 26(18), 9056; https://doi.org/10.3390/ijms26189056 - 17 Sep 2025
Viewed by 490
Abstract
Single three-lactic acid bacterial strains with anti-adipogenic effects in C3H10T1/2 cells and possessing beneficial probiotic properties were administered to mice fed a high-fat diet. Of the three strains, Lactiplantibacillus plantarum RP12, which had the lowest weight gain, was utilized for further studies, including [...] Read more.
Single three-lactic acid bacterial strains with anti-adipogenic effects in C3H10T1/2 cells and possessing beneficial probiotic properties were administered to mice fed a high-fat diet. Of the three strains, Lactiplantibacillus plantarum RP12, which had the lowest weight gain, was utilized for further studies, including a second mouse experiment lasting 10 weeks. Oral administration of Lactiplantibacillus plantarum RP12 resulted in reduced body weight gain and epididymal fat mass. Significant reductions in serum total cholesterol, triglycerides, and blood glucose were observed in the group treated with Lactiplantibacillus plantarum RP12. This strain was found to regulate the expression of genes associated with lipid metabolism in epididymal adipose tissue and liver. It induced changes in the composition of fecal microbiota. Although there is no difference in the Bacillota to Bacteroidota ratio between the HFD and RP12 groups, notable differences in the compositions at the family, genus, and species levels were evident. Specifically, differences in the proportions of some taxa reported to have an association with obesity were observed between the HFD and RP12 groups. Fecal analyses demonstrated that Lactiplantibacillus plantarum RP12 diminishes lipid absorption and augments the production of short-chain fatty acids in the intestine. Lactiplantibacillus plantarum RP12 also mitigated damage to the morphology of the ileum and colon caused by a high-fat diet and promoted the expression of Claudin-1 and Muc2. Overall, Lactiplantibacillus plantarum RP12 has potential as a useful probiotic to address metabolic disorders as well as obesity, substantiating the positive in vivo indicators and modulation of gut microbiota in a high-fat diet-induced obese mouse model. Full article
(This article belongs to the Section Molecular Microbiology)
Show Figures

Figure 1

22 pages, 5476 KB  
Article
Impact of High-Fat Diet and Aflatoxin B1 on Immunometabolic Dysfunction and the Dose-Responsive Modulation by Isoleucine Supplementation
by Ruojin Wang, Jiangli Wang, Meifang Lan and Xiyin Wang
Nutrients 2025, 17(17), 2897; https://doi.org/10.3390/nu17172897 - 8 Sep 2025
Viewed by 669
Abstract
Objectives: Disruption of gut–liver axis homeostasis is a hallmark of metabolic and toxic stress. This study aimed to evaluate the combined effects of high-fat diet (HFD), aflatoxin B1 (AFB1), and exogenous isoleucine supplementation on immunometabolic function under nutritional and toxic stress. Methods: [...] Read more.
Objectives: Disruption of gut–liver axis homeostasis is a hallmark of metabolic and toxic stress. This study aimed to evaluate the combined effects of high-fat diet (HFD), aflatoxin B1 (AFB1), and exogenous isoleucine supplementation on immunometabolic function under nutritional and toxic stress. Methods: Two-phase murine experiments assessed: (1) HFD and AFB1 effects individually and combined; and (2) dose-dependent isoleucine responses (25/50/100 mg/kg) across control, HFD, and HFD + AFB1 backgrounds. Results: HFD significantly impaired liver function, promoted Th17-mediated inflammation, and induced gut dysbiosis, while AFB1 alone exerted minimal effects. Their combination synergistically exacerbated hepatic steatosis, intestinal barrier disruption, and inflammatory responses. Fecal metabolomics identified elevated isoleucine as a potential inflammatory biomarker. Under HFD, isoleucine (50 mg/kg) amplified inflammation and oxidative stress. Remarkably, under HFD + AFB1, moderate/high-dose isoleucine reduced hepatic lipid deposition and triglycerides despite persistent intestinal damage, demonstrating context-dependent effects. Conclusions: HFD and AFB1 synergistically disrupt gut–liver axis integrity through immunometabolic mechanisms. Isoleucine supplementation exhibits dual-modulatory effects, exacerbating damage under nutritional stress while partially mitigating hepatic lipid accumulation under combined toxic-nutritional stress, highlighting the critical importance of environmental context in amino acid interventions. Full article
(This article belongs to the Special Issue Health Effects of Diet-Sourced Hazardous Factors)
Show Figures

Figure 1

17 pages, 10116 KB  
Article
Blueberry Anthocyanins Ameliorate Hepatic Dysfunction in High-Fat Diet-Fed Mice: Association with Altered Gut Microbiota and Bile Acid Metabolism
by Peng Xu, Yucheng He, Junyi Wang, Yingkun Sheng and Jianfeng Wang
Foods 2025, 14(17), 3121; https://doi.org/10.3390/foods14173121 - 6 Sep 2025
Viewed by 729
Abstract
The rapid rise in obesity has evolved into a critical global public health concern. Considering the potential adverse effects of current anti-obesity medications, the development of functional foods sourced from natural materials has emerged as a viable alternative. Blueberries, a category of berry [...] Read more.
The rapid rise in obesity has evolved into a critical global public health concern. Considering the potential adverse effects of current anti-obesity medications, the development of functional foods sourced from natural materials has emerged as a viable alternative. Blueberries, a category of berry fruits, exhibit potential anti-obesity characteristics. In this research, we assessed the impacts of Blueberry extract rich in anthocyanins (BE) on lipid metabolism and liver health in a high-fat diet (HFD)-induced obese mouse model. The findings indicated that BE notably diminished lipid accumulation in both serum and the liver, and mitigated hepatic steatosis and oxidative stress. Integrated proteomic, metagenomic, and metabolomic analyses further revealed the underlying mechanisms. Consumption of BE intake reconfigured the gut microbiota composition and reduced the microbial capacity for secondary bile acid metabolism, thereby interrupting bile acid recycling and facilitating fecal excretion. This process led to a reduction in systemic cholesterol levels and ultimately alleviated hepatic lipid accumulation, resulting in enhanced liver health. Full article
(This article belongs to the Section Nutraceuticals, Functional Foods, and Novel Foods)
Show Figures

Figure 1

42 pages, 1227 KB  
Review
Microbial Metabolomes in Alzheimer’s Disease: From Pathogenesis to Therapeutic Potential
by Alejandro Borrego-Ruiz and Juan J. Borrego
Curr. Issues Mol. Biol. 2025, 47(9), 724; https://doi.org/10.3390/cimb47090724 - 5 Sep 2025
Viewed by 835
Abstract
Background: Accumulating evidence underscores the potential role of the gut microbiome in the pathogenesis of Alzheimer’s disease, but much remains to be clarified. This review examines current evidence linking gut microbiome dysbiosis to Alzheimer’s disease, focusing on microbial metabolomes and their mechanistic role, [...] Read more.
Background: Accumulating evidence underscores the potential role of the gut microbiome in the pathogenesis of Alzheimer’s disease, but much remains to be clarified. This review examines current evidence linking gut microbiome dysbiosis to Alzheimer’s disease, focusing on microbial metabolomes and their mechanistic role, as well as on the potential of therapeutic approaches targeting the gut microbiome. Methods: A narrative, non-systematic examination of the literature was conducted to provide a comprehensive overview of the subject under examination. Database searches were performed in PubMed, Scopus, and Web of Science between June and July 2025. Results: Alzheimer’s disease is linked to reduced gut microbial diversity and altered bacterial taxa. Gut microbiome shifts correlate with inflammation and may drive Alzheimer’s disease progression via the microbiota–gut–brain axis. Microbial amyloids and bacterial products can cross both the intestinal and blood–brain barrier, triggering neuroinflammation and promoting amyloid and tau pathologies. Short-chain fatty acids produced by the gut microbiome regulate neuroinflammation, lipid metabolism, and gene expression, impacting Alzheimer’s disease pathology. Therapeutics targeting the gut microbiome, including probiotics, prebiotics, and fecal microbiota transplantation, show promise in modulating neuroinflammation, reducing amyloid and tau pathology, and improving cognitive function in Alzheimer’s disease. Conclusions: The gut microbiome significantly influences Alzheimer’s disease pathogenesis, and its modulation offers potential to slow progression. However, further research is required to validate effective clinical interventions. Full article
Show Figures

Figure 1

32 pages, 2106 KB  
Review
Gut Microbiota-Derived Metabolites in Atherosclerosis: Pathways, Biomarkers, and Targets
by Alexandra-Kristine Tonch-Cerbu, Adrian-Gheorghe Boicean, Oana-Maria Stoia and Minodora Teodoru
Int. J. Mol. Sci. 2025, 26(17), 8488; https://doi.org/10.3390/ijms26178488 - 1 Sep 2025
Cited by 1 | Viewed by 1392
Abstract
The human gut microbiota is a complex ecosystem that influences host metabolism, immune function, and cardiovascular health. Dysbiosis, defined as an imbalance in microbial composition or function, has been linked to the development and progression of atherosclerosis. This connection is mediated by microbial [...] Read more.
The human gut microbiota is a complex ecosystem that influences host metabolism, immune function, and cardiovascular health. Dysbiosis, defined as an imbalance in microbial composition or function, has been linked to the development and progression of atherosclerosis. This connection is mediated by microbial metabolites that enter the systemic circulation and interact with vascular and immune pathways. Among these, trimethylamine N-oxide (TMAO) has been most extensively studied and is consistently associated with cardiovascular events. Other metabolites, including lipopolysaccharides (LPS), short-chain fatty acids (SCFAs), and secondary bile acids, also contribute by modulating inflammation, endothelial function, and lipid metabolism. Recent research has expanded to emerging metabolites such as indoxyl sulfate, indole-3-propionic acid, and polyamines, which may provide additional mechanistic insights. These microbial products are increasingly explored as biomarkers of cardiovascular risk. TMAO has shown predictive value in large human cohorts, while microbiota composition and diversity measures remain less consistent across studies. However, interpretation of these biomarkers is limited by methodological variability, interindividual differences, and lack of standardization. Therapeutic interventions targeting the gut–heart axis are under investigation. Dietary strategies such as the Mediterranean diet and fiber-rich nutrition, probiotics and prebiotics, and fecal microbiota transplantation (FMT) show promise, while pharmacological approaches targeting TMAO or bile acid pathways are in early stages. This review summarizes current knowledge on the mechanistic, diagnostic, and therapeutic links between the gut microbiota and atherosclerosis, highlighting both established findings and emerging directions for future research. Full article
(This article belongs to the Special Issue Cellular and Molecular Progression of Cardiovascular Diseases)
Show Figures

Figure 1

15 pages, 5483 KB  
Article
Modulation of the Gut Microbiota by Nopalea cochenillifera (Prickly Pear Cactus) Contributes to Improved Lipid Metabolism and Immune Function
by Sayaka Yokoyama, Amane Kikuchi, Hideaki Takahashi, Hinako Ushimaru, Hibiki Yamaguchi, Chikako Yamada, Kotoyo Fujiki, Hana Kozai, Suzuno Ota, Tadashi Fujii, Yoshiki Hirooka, Takumi Tochio and Mamoru Tanaka
Nutrients 2025, 17(17), 2844; https://doi.org/10.3390/nu17172844 - 31 Aug 2025
Viewed by 1180
Abstract
Background/Objectives: Nopalea cochenillifera (L.) Salm-Dyck cladodes are rich in dietary fiber, polyphenols, and minerals, which are known to exert antioxidant and immunomodulatory effects. However, the mechanisms and active constituents have not been fully elucidated. In this study, we investigated the effects of [...] Read more.
Background/Objectives: Nopalea cochenillifera (L.) Salm-Dyck cladodes are rich in dietary fiber, polyphenols, and minerals, which are known to exert antioxidant and immunomodulatory effects. However, the mechanisms and active constituents have not been fully elucidated. In this study, we investigated the effects of continuous N. cochenillifera consumption on lipid metabolism, immune function, and the gut microbiota in mice. Methods: The feed was made using freeze-dried and powdered cladodes of N. cochenillifera. Male C57BL/6J mice were assigned to four groups: control diet (C), control diet plus 10% N. cochenillifera (CN), high-fat diet (FC), and high-fat diet plus 10% N. cochenillifera (FN). Results: Cactus supplementation reduced the body and liver weights that were elevated by the high-fat diet. Serum total cholesterol and free fatty acids were increased in the FC group compared with the C group, while cactus intake lowered these levels and enhanced fecal cholesterol excretion. Cactus consumption also elevated fecal total IgA and mucin contents. IL-4 expression in Peyer’s patches was significantly increased in the FN group compared with the FC group. Gut microbiota analysis showed significant differences in β-diversity, along with increased α-diversity and higher abundance of Lachnospiraceae, following cactus intake. Conclusions: These findings suggest that N. cochenillifera intake increases gut microbiota diversity, which enhances intestinal barrier function and thereby contributes to improved lipid metabolism and immune regulation. Full article
(This article belongs to the Special Issue Functional Foods and Sustainable Health (2nd Edition))
Show Figures

Figure 1

22 pages, 7389 KB  
Article
Mangosteen Pericarp Extract Mitigates Diquat-Induced Hepatic Oxidative Stress by NRF2/HO-1 Activation, Intestinal Barrier Integrity Restoration, and Gut Microbiota Modulation
by Weichen Huang, Yujie Lv, Chenhao Zou, Chaoyue Ge, Shenao Zhan, Xinyu Shen, Lianchi Wu, Xiaoxu Wang, Hongmeng Yuan, Gang Lin, Dongyou Yu and Bing Liu
Antioxidants 2025, 14(9), 1045; https://doi.org/10.3390/antiox14091045 - 25 Aug 2025
Viewed by 863
Abstract
Poultry production exposes birds to diverse environmental and physiological stressors that disrupt redox balance, impair gut–liver axis function, and undermine health and productivity. This study investigated the hepatoprotective and antioxidative effects of mangosteen pericarp extract (MPE) in an experimental model of diquat-induced oxidative [...] Read more.
Poultry production exposes birds to diverse environmental and physiological stressors that disrupt redox balance, impair gut–liver axis function, and undermine health and productivity. This study investigated the hepatoprotective and antioxidative effects of mangosteen pericarp extract (MPE) in an experimental model of diquat-induced oxidative stress in laying hens. A total of 270 Hy-Line White laying hens were randomly assigned to three groups: control group (CON), diquat-challenged group (DQ), and MEP intervention with diquat-challenged group (MQ), with six replicates of 15 birds each. The results showed that MPE supplementation effectively mitigated the hepatic oxidative damage caused by diquat, as evidenced by the increased ALT and AST activity, improved lipid metabolism, and reduced hepatic fibrosis. Mechanistically, MPE activated the NRF2/HO-1 antioxidant pathway, thus enhancing the liver’s ability to counteract ROS-induced damage and reducing lipid droplet accumulation in liver tissue. MPE supplementation restored intestinal barrier integrity by upregulating tight junction protein expression (Occludin-1 and ZO-1), enhancing MUC-2 expression, and thereby decreasing gut microbiota-derived LPS transferring from the intestine. Additionally, MPE also modulated gut microbiota composition by enriching beneficial bacterial genera such as Lactobacillus and Ruminococcus while suppressing the growth of potentially harmful taxa (e.g., Bacteroidales and UCG-010). Fecal microbiota transplantation (FMT) from MPE-treated donors into diquat-exposed recipients reproduced these beneficial effects, further highlighting the role of gut microbiota modulation in mediating MPE’s systemic protective actions. Together, these findings demonstrated that MPE alleviated DQ-induced liver injury and oxidative stress through a combination of antioxidant activity, protection of intestinal barrier function, and modulation of gut microbiota, positioning MPE as a promising natural strategy for mitigating oxidative stress-related liver damage by regulating the gut microbiota and gut–liver axis in poultry. Full article
(This article belongs to the Special Issue Oxidative Stress in Animal Reproduction and Nutrition)
Show Figures

Graphical abstract

16 pages, 2491 KB  
Article
Gut Microbiota Modulation and Anti-Obesity Potential of Epigallocatechin-3-Gallate-Quercetin-Rutin Against High-Fat Diet-Induced Obesity in Rats
by Yu-Jou Chien, Ching-Chang Cho, Yu-Ting Hung, Li-You Chen, Yue-Ching Wong, Shiuan-Chih Chen and Chin-Lin Hsu
Life 2025, 15(8), 1331; https://doi.org/10.3390/life15081331 - 21 Aug 2025
Viewed by 863
Abstract
Polyphenols have been widely recognized for their potential anti-obesity effects. This study aimed to evaluate the impact of a polyphenol compound-epigallocatechin-3-gallate, quercetin, and rutin (EQR) on obesity-related parameters and gut microbiota composition. After four weeks of high-fat diet (HFD) induction, the obese Wistar [...] Read more.
Polyphenols have been widely recognized for their potential anti-obesity effects. This study aimed to evaluate the impact of a polyphenol compound-epigallocatechin-3-gallate, quercetin, and rutin (EQR) on obesity-related parameters and gut microbiota composition. After four weeks of high-fat diet (HFD) induction, the obese Wistar male rats received EQR treatment for an additional four weeks. EQR supplementation significantly reduced body weight gain, feed efficiency, adipose tissue accumulation, and liver lipid content in obese rats. Additionally, it enhanced fecal short-chain fatty acid (SCFA) levels and modulated gut microbiota composition. Specifically, EQR treatment significantly induced Fusobacteria, Fusobacteriaceae, Christensenellaceae, Christensenellaceae R-7 group, Lachnoclostridium, Enterorhabdus, and Parvibacter levels and reduced Deferribacteres and Mucispirillum levels. Gene expression analysis in liver, white adipose tissue (WAT), and brown adipose tissue (BAT) revealed that EQR upregulated the expression of liver PPAR-α, WAT SIRT-1, and BAT PGC-1α, while downregulating liver PPAR-γ, liver FATP-1, and WAT FAS, indicating its role in promoting fatty acid oxidation and thermogenesis, as well as suppressing lipid synthesis and transport. In conclusion, EQR demonstrated significant anti-obesity effects by modulating gut microbiota and lipid metabolism, suggesting its potential as a functional ingredient for obesity management. Full article
Show Figures

Figure 1

17 pages, 26824 KB  
Article
Honey-Conjugated Honeybee Brood Biopeptides Improve Gastrointestinal Stability, Antioxidant Capacity, and Alleviate Diet-Induced Metabolic Syndrome in a Rat Model
by Sakaewan Ounjaijean, Supakit Chaipoot, Rewat Phongphisutthinant, Gochakorn Kanthakat, Sirinya Taya, Pattavara Pathomrungsiyounggul, Pairote Wiriyacharee and Kongsak Boonyapranai
Foods 2025, 14(16), 2907; https://doi.org/10.3390/foods14162907 - 21 Aug 2025
Viewed by 552
Abstract
Honeybee brood biopeptides (HBb-Bps) are a novel source of bioactive compounds with potential health benefits. In this study, HBb-Bps were conjugated with honey via a Maillard reaction and their physicochemical properties, digestive stability, antioxidant capacity, and anti-obesogenic effects were evaluated. Simulated gastrointestinal digestion [...] Read more.
Honeybee brood biopeptides (HBb-Bps) are a novel source of bioactive compounds with potential health benefits. In this study, HBb-Bps were conjugated with honey via a Maillard reaction and their physicochemical properties, digestive stability, antioxidant capacity, and anti-obesogenic effects were evaluated. Simulated gastrointestinal digestion revealed significantly enhanced resistance after conjugation, with the residual content increasing from 46.99% for native HBb-Bps to 86.12% for the honey-conjugated forms; furthermore, antioxidant activity was largely preserved according to the DPPH and ABTS assays. In the in vivo experiments, 30 male BrlHan: WIST@Jcl (GALAS) (Wistar) rats were fed a high-fat diet (HFD) to induce obesity and orally administered honey-conjugated HBb-Bps at doses of 200, 500, or 1000 mg/kg body weight for 16 weeks. The highest dose led to significant reductions in body weight gain, the Lee index, and body mass index. The serum lipid profiles markedly improved, with decreases in the total cholesterol, triglyceride, and LDL levels, as well as cardiovascular risk indices. Furthermore, fecal analysis showed increased levels of short-chain fatty acids, particularly butyrate. These changes suggest enhanced gut microbial activity; however, the prebiotic effects were inferred from the SCFA profiles, as the gut microbiota composition was not directly analyzed. In conclusion, honey-conjugated HBb-Bps improve gastrointestinal stability and exhibit antioxidant, hypolipidemic, and gut-modulating effects, supporting their potential use as functional ingredients for managing diet-induced metabolic disorders. Full article
Show Figures

Figure 1

24 pages, 6521 KB  
Article
Liubao Tea Extract Attenuates High-Fat Diet and Streptozotocin-Induced Type 2 Diabetes in Mice by Remodeling Hepatic Metabolism and Gut Microbiota
by Jichu Luo, Zhijuan Wei, Yuru Tan, Ying Tong, Bao Yang, Mingsen Wen, Xuan Guan, Pingchuan Zhu, Song Xu, Xueting Lin and Qisong Zhang
Nutrients 2025, 17(16), 2665; https://doi.org/10.3390/nu17162665 - 18 Aug 2025
Viewed by 1152
Abstract
Background: Type 2 diabetes (T2D) has become a serious global public health concern. Liubao tea (LBT) has demonstrated beneficial effects on gut microbiota and glucose-lipid metabolism, holding promising therapeutic potential for T2D; however, its underlying mechanisms remain unclear. This study aims to [...] Read more.
Background: Type 2 diabetes (T2D) has become a serious global public health concern. Liubao tea (LBT) has demonstrated beneficial effects on gut microbiota and glucose-lipid metabolism, holding promising therapeutic potential for T2D; however, its underlying mechanisms remain unclear. This study aims to elucidate the potential mechanisms of Liubao tea extract (LBTE) against T2D. Methods: LC-MS technology was used to identify the chemical components of LBTE and combined with network pharmacology and molecular docking to screen its potential active ingredients and targets for improving T2D. Therapeutic efficacy was assessed in high-fat diet/streptozotocin (HFD/STZ)-induced diabetic mice via serum biochemical analyses and histopathological examinations. Serum metabolomics, 16S rRNA sequencing, quantification of short-chain fatty acids (SCFAs), quantitative real-time PCR (qPCR), and antibiotic-treated pseudo-germ-free models were employed to elucidate the underlying mechanisms. Results: LBTE effectively reduced blood glucose levels and improved lipid metabolism, primarily by promoting hepatic glycogen synthesis and suppressing glycerophospholipid synthesis. LBTE also alleviated hepatic inflammation by modulating inflammatory cytokine expression. Additionally, LBTE reshaped the gut microbiota profiles by decreasing harmful bacteria and increasing SCFA-producing bacteria, resulting in elevated fecal SCFAs. SCFAs contributed to improving hepatic metabolism and inflammation, enhancing intestinal barrier function. Notably, these effects were abolished by antibiotic-induced microbiota depletion, confirming the microbiota-dependent mechanism of LBTE. Quercetin, luteolin, genistein, and kaempferol were considered as potential active ingredients contributing to the antidiabetic effects of LBTE. Conclusions: These findings provide novel perspectives on the viability of LBTE as a complementary strategy for T2D prevention and management. Full article
(This article belongs to the Section Phytochemicals and Human Health)
Show Figures

Figure 1

22 pages, 3707 KB  
Article
Gut–Liver Axis-Mediated Anti-Obesity Effects and Viscosity Characterization of a Homogenized Viscous Vegetable Mixture in Mice Fed a High-Fat Diet
by Yu-An Wei, Yi-Hsiu Chen, Lu-Chi Fu, Chiu-Li Yeh, Shyh-Hsiang Lin, Yuh-Ting Huang, Yasuo Watanabe and Suh-Ching Yang
Plants 2025, 14(16), 2510; https://doi.org/10.3390/plants14162510 - 12 Aug 2025
Viewed by 717
Abstract
This study investigated the anti-obesity effects of a homogenized, viscous vegetable (VV) mixture prepared from mucilaginous vegetables, with a focus on modulating hepatic lipid metabolism and gut microbiota composition in mice fed with a high-fat (HF) diet. The VV mixture was formulated by [...] Read more.
This study investigated the anti-obesity effects of a homogenized, viscous vegetable (VV) mixture prepared from mucilaginous vegetables, with a focus on modulating hepatic lipid metabolism and gut microbiota composition in mice fed with a high-fat (HF) diet. The VV mixture was formulated by blending freeze-dried powders of ten mucilaginous vegetables, classified as moderately thick using a line-spread test and extremely thick according to the IDDSI framework in a 1:9 ratio (VV mixture: water, w/w). Six-week-old male C57BL/6 mice were fed control or HF diets, with or without 10% VV mixture for 8 weeks (n = 7 per group). The HF diet induced significant weight gain, adipose tissue accumulation, hepatic steatosis, and inflammation. The HF diet also significantly reduced hepatic ACO1, CPT1 mRNA expression, and α-diversity with distinct fecal microbiota profiles. On the other hand, VV mixture supplementation reduced serum TC, LDL-C levels and NAFLD scores. VV mixture supplementation also increased hepatic ACO1 and CPT1 mRNA expression, enhanced α-diversity, and enriched SCFA-producing bacteria, particularly the Lachnospiraceae NK4A136 group. In conclusion, the VV mixture attenuated HF diet-induced obesity, possibly through its high viscosity–mediated effects on hepatic fatty acid oxidation and gut microbiota modulation. Full article
(This article belongs to the Section Phytochemistry)
Show Figures

Graphical abstract

15 pages, 1474 KB  
Article
Decline in Serum Lysophosphatidylcholine Species in Patients with Severe Inflammatory Bowel Disease
by Hauke Christian Tews, Tanja Elger, Muriel Huss, Johanna Loibl, Arne Kandulski, Martina Müller, Marcus Höring, Gerhard Liebisch and Christa Buechler
J. Clin. Med. 2025, 14(15), 5485; https://doi.org/10.3390/jcm14155485 - 4 Aug 2025
Viewed by 687
Abstract
Background/Objectives: Lysophosphatidylcholine (LPC) is composed of various lipid species, some of which exert pro-inflammatory and others anti-inflammatory activities. However, most of the LPC species analyzed to date are reduced in the serum of patients with inflammatory bowel disease (IBD) compared to healthy [...] Read more.
Background/Objectives: Lysophosphatidylcholine (LPC) is composed of various lipid species, some of which exert pro-inflammatory and others anti-inflammatory activities. However, most of the LPC species analyzed to date are reduced in the serum of patients with inflammatory bowel disease (IBD) compared to healthy controls. To our knowledge, the correlation between serum LPC species levels and measures of inflammation, as well as their potential as markers for monitoring IBD activity, has not yet been investigated. Methods: Thirteen LPC species, varying in acyl chain length and number of double bonds, were measured in the serum of 16 controls and the serum of 57 patients with IBD. Associations with C-reactive protein (CRP) and fecal calprotectin levels as markers of IBD severity were assessed. Results: Serum levels of LPC species did not differ between the healthy controls and the entire patient cohort. In patients with IBD, serum levels of LPC 16:1, 18:0, 18:3, 20:3, and 20:5, as well as total LPC concentrations, showed inverse correlations with both CRP and fecal calprotectin levels, indicating an association with inflammatory activity. Nine LPC species were significantly reduced in patients with high fecal calprotectin compared to those with low values. LPC species with 22 carbon atoms and 4 to 6 double bonds were not related to disease activity. Stool consistency and gastrointestinal symptoms did not influence serum LPC profiles. Corticosteroid treatment was associated with lower serum LPC 20:3 and 22:5 levels, while mesalazine, anti-TNF, and anti-IL-12/23 therapies had no significant impact on LPC concentrations. There was a strong positive correlation between LPC species containing 15 to 18 carbon atoms and serum cholesterol, triglycerides, and phosphatidylcholine levels. However, there was no correlation with markers of liver disease. Conclusions: Shorter-chain LPC species are reduced in patients with active IBD and reflect underlying hypolipidemia. While these lipid alterations provide insight into IBD-associated metabolic changes, they appear unsuitable as diagnostic or disease monitoring biomarkers. Full article
(This article belongs to the Special Issue Inflammatory Bowel Disease: Pathogenesis and Management Strategies)
Show Figures

Figure 1

15 pages, 1125 KB  
Article
Mixed Green Banana (Musa spp.) Pulp and Peel Flour Reduced Body Weight Gain and Adiposity and Improved Lipid Profile and Intestinal Morphology in Wistar Rats
by Leonara Martins Viana, Bárbara Pereira da Silva, Fabiana Silva Rocha Rodrigues, Laise Trindade Paes, Marcella Duarte Villas Mishima, Renata Celi Lopes Toledo, Elad Tako, Hércia Stampini Duarte Martino and Frederico Barros
Nutrients 2025, 17(15), 2493; https://doi.org/10.3390/nu17152493 - 30 Jul 2025
Viewed by 868
Abstract
Background and Objectives: In recent years, there has been growing interest in the production of ingredients rich in dietary fiber and antioxidants, such as green banana flours. This study evaluated the effect of consumption of mixed green banana pulp (PF) and peel (PeF) [...] Read more.
Background and Objectives: In recent years, there has been growing interest in the production of ingredients rich in dietary fiber and antioxidants, such as green banana flours. This study evaluated the effect of consumption of mixed green banana pulp (PF) and peel (PeF) flours on the body weight gain, adiposity, lipid profile, and intestinal morphology of Wistar rats. Methods: Male young rats were divided into four groups (n = 8) that received a standard diet (SD), or one of the following three test diets: M1 (SD + 90% PF/10% PeF), M2 (SD + 80% PF/20% PeF), or P (SD + 100% PF) for 28 days. Results: Rats from M1, M2, and P groups showed reduced body weight gain and adiposity and had lower contents of total cholesterol, LDL-c, VLDL-c, and triglycerides. Animals from M1 and M2 groups had an increase in cecum weight, fecal moisture, acetic acid concentration, and crypt depth and reduced fecal pH. Moreover, consumption of the M1, M2, and P diets increased the expression of proteins involved in intestinal functionality. Significant negative correlations were observed between consumption of resistant starch and soluble dietary fiber, from the flours, and weight gain (r = −0.538 and r = −0.538, respectively), body adiposity (r = −0.780 and r = −0.767, respectively), total cholesterol (r = −0.789 and r = −0.800, respectively), and triglycerides (r = −0.790 and r = −0.786, respectively). Conclusions: Mixed green banana pulp and peel flour proved to be a viable alternative as a food ingredient that can promote weight loss, improve lipid profile and intestinal morphology, and minimize post-harvest losses. Full article
(This article belongs to the Section Nutrition and Obesity)
Show Figures

Figure 1

Back to TopTop