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

Search Results (210)

Search Parameters:
Keywords = hepatic carbohydrate metabolism

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
24 pages, 1139 KB  
Review
SLC35 Transporters: The Missing Link Between Glycosylation and Type 2 Diabetes
by Xu Zhang, Hafiza Mahreen Mehwish and Pulin Che
Diabetology 2026, 7(1), 7; https://doi.org/10.3390/diabetology7010007 - 1 Jan 2026
Viewed by 287
Abstract
Type 2 diabetes mellitus (T2D) affects hundreds of millions worldwide, with recent estimates indicating approximately 589 million adults living with diabetes, most with type 2 disease. Beyond classical insulin signaling pathways, increasing evidence implicates altered protein glycosylation in metabolic dysfunction. The solute carrier [...] Read more.
Type 2 diabetes mellitus (T2D) affects hundreds of millions worldwide, with recent estimates indicating approximately 589 million adults living with diabetes, most with type 2 disease. Beyond classical insulin signaling pathways, increasing evidence implicates altered protein glycosylation in metabolic dysfunction. The solute carrier 35 (SLC35) family of nucleotide sugar transporters mediates the import of activated sugars into the endoplasmic reticulum and Golgi lumen, thereby influencing global glycosylation patterns. Dysregulation of these transporters can perturb glucose homeostasis, insulin responsiveness, and nutrient-sensing pathways through changes in glycosylation flux. In this review, we dissect the molecular mechanisms by which these transporters modulate glucose homeostasis, insulin signaling pathways, protein O-GlcN acylation, and broader glycosylation processes. We integrate findings from human genetic studies, rodent models, and in vitro functional analyses to characterize how altered SLC35 activity is associated with T2D and metabolic syndrome. Four members demonstrate particularly compelling evidence: SLC35B4 modulates hepatic glucose metabolism, SLC35D3 mutations impair dopaminergic signaling and energy balance, and SLC35F3 variants interact with high-carbohydrate intake to increase metabolic-syndrome risk. SLC35A3, though less studied, may influence glycosylation-dependent insulin signaling through its role in N-glycan biosynthesis. Beyond these characterized transporters, this review identifies potential metabolic roles for understudied family members, suggesting broader implications across the entire SLC35 family. We also discuss how such alterations can lead to disrupted hexosamine flux, impaired glycoprotein processing, aberrant cellular signaling, and micronutrient imbalances. Finally, we evaluate the therapeutic potential of targeting SLC35 transporters, outlining both opportunities and challenges in translating these insights into novel T2D treatments. Full article
(This article belongs to the Special Issue New Perspectives on Diabetes and Stroke Research)
Show Figures

Figure 1

19 pages, 4713 KB  
Article
Bacillus subtilis FZU103 Promotes Growth in Micropterus salmoides, Accompanied by Modulation of Gut Microbiota, Enhanced Liver Antioxidants and Digestive Enzyme Activity
by Xu Chen, Hong Zheng, Wenrui Liang, Yinggu Kuang, Xiangzhu Shi, Jinlin Fan, Xucong Lv and Jiacong Deng
Microorganisms 2026, 14(1), 93; https://doi.org/10.3390/microorganisms14010093 - 31 Dec 2025
Viewed by 241
Abstract
Probiotics hold great potential in aquaculture, as they can effectively modulate gut microbiota and improve fish health, thereby enhancing farming efficiency. Translating this potential into practical application critically relies on screening high-efficacy probiotic strains. This study evaluated the growth-promoting and health-enhancing effects of [...] Read more.
Probiotics hold great potential in aquaculture, as they can effectively modulate gut microbiota and improve fish health, thereby enhancing farming efficiency. Translating this potential into practical application critically relies on screening high-efficacy probiotic strains. This study evaluated the growth-promoting and health-enhancing effects of probiotic candidates Lactobacillus rhamnosus GG (LGG), Lactobacillus plantarum FZU310 (LP-FZU310) and Bacillus subtilis FZU103 (BS-FZU103) in largemouth bass (Micropterus salmoides). After feeding different probiotics for 30 days, the growth, antioxidant, and intestinal enzyme indicators of M. salmoides were detected. BS-FZU103 demonstrated superior efficacy among the tested strains, showing significant differences in both specific growth rate (SGR) (p < 0.05) and condition factor (CF) (p < 0.05). It also markedly enhanced hepatic antioxidant status, elevating superoxide dismutase and glutathione peroxidase activities while reducing malondialdehyde levels by 80%. Improved liver integrity was indicated by significant decreases in serum alanine aminotransferase, aspartate aminotransferase and alkaline phosphatase. Digestively, BS-FZU103 specifically increased intestinal amylase activity by 14.7%, without affecting protease or lipase, suggesting enhanced carbohydrate digestion. 16S rRNA sequencing revealed BS-FZU103 remodeled gut microbiota, increasing Proteobacteria abundance at the phylum level and enriching Bacillus while reducing Clostridium sensu stricto 1 at the genus level. Functional prediction based on PICRUSt2 indicated an enhanced metabolic potential of the gut microbiota, with inferred upregulation of pathways related to carbohydrate transport and metabolism (e.g., ABC transporters) and intestinal enzymatic activities. Collectively, BS-FZU103 is associated with metabolic modulation, promoting M. salmoides growth through gut microbiota remodeling, hepatic antioxidant fortification, and targeted augmentation of carbohydrate utilization efficiency. Full article
(This article belongs to the Section Gut Microbiota)
Show Figures

Figure 1

12 pages, 913 KB  
Review
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): New Perspectives on an Evolving Epidemic
by Gerond Lake-Bakaar
J. Clin. Med. 2025, 14(24), 8872; https://doi.org/10.3390/jcm14248872 - 15 Dec 2025
Viewed by 603
Abstract
The absence of a unifying pathogenetic mechanism in metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), has significantly hindered therapeutic progress. Appreciation that the delivery of excessive amounts of calories to the liver via the portal circulation [...] Read more.
The absence of a unifying pathogenetic mechanism in metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), has significantly hindered therapeutic progress. Appreciation that the delivery of excessive amounts of calories to the liver via the portal circulation might be a key parallel between MASLD and the twin steatotic liver disease, alcohol-related liver disease (ALD), establishes a consolidated framework that could guide rational drug design and precise therapeutic approaches. This review contends that, in both ALD and MASLD, the unique dual blood supply to the liver, from both portal vein and hepatic artery as well as the distinctive blood flow control physiology, prevents hepatic arterial oxygen delivery from adequately compensating for the increased metabolic demands induced by excess caloric intake—alcohol in ALD and food in MASLD—resulting in hepatocellular injury. Over four decades ago, Lautt postulated that this ‘oxygen-nutrient mismatch’ could play a role in ALD. We have extended this paradigm to MASLD, theorizing that analogous mechanisms may be involved in both conditions. Evidence that comorbidities, which are associated with recurrent episodes of hypoxemia, such as obstructive sleep apnea (OSA), exacerbate MASLD progression, supports this. ALD is less strongly linked to metabolic syndrome than MASLD. This may be due to inherent differences in hepatic substrate processing. Carbohydrates, lipids, and proteins undergo diverse and flexible cytosolic metabolic pathways, especially under metabolic stress. In contrast, hepatic ethanol metabolism is predominantly linear and obligately oxidative, providing limited metabolic adaptability. Future perspectives could focus on rectifying the imbalance between hepatic oxygen delivery and nutrient availability. This might be accomplished by attenuating hepatic caloric excess using emerging pharmacotherapies for weight reduction, augmenting hepatic oxygenation through hyperbaric oxygen therapy, or increasing hepatic arterial blood flow with agents such as obeticholic acid. Furthermore, enhancement of hepatic basal metabolic activity with thyroid hormone receptor-β agonists, like resmiritom may confer similar therapeutic effects. Full article
Show Figures

Figure 1

17 pages, 7083 KB  
Article
Cassava Fiber Prevents High-Fat Diet-Induced Obesity in Mice Through Gut Microbiota Restructuring
by Yajin Yang, Fuhong Lei, Lily Liu, Yanhong Chen, Qingqing Li, Jieming Long, Zubing Zhang and Aiwei Guo
Foods 2025, 14(23), 4121; https://doi.org/10.3390/foods14234121 - 1 Dec 2025
Viewed by 666
Abstract
Cassava fiber (CF) is a novel dietary fiber extracted from cassava by-products. To investigate its anti-obesity mechanism, obesity was induced in mice through a high-fat diet (HFD). Dietary supplementation with 10% CF significantly reduced body weight, body fat, triglycerides, low-density lipoprotein cholesterol, total [...] Read more.
Cassava fiber (CF) is a novel dietary fiber extracted from cassava by-products. To investigate its anti-obesity mechanism, obesity was induced in mice through a high-fat diet (HFD). Dietary supplementation with 10% CF significantly reduced body weight, body fat, triglycerides, low-density lipoprotein cholesterol, total cholesterol, and fasting blood glucose in mice. CF effectively ameliorated hepatic steatosis and adipocyte hypertrophy, increased the villus height-to-crypt depth ratio, enhanced mucus secretion by intestinal goblet cells, down-regulated the expression of ileal lipid absorption-related genes (NPC1L1, CD36, and FABP2), and up-regulated the short-chain fatty acid receptor GPR43, collectively improving intestinal health. Compared to HFD mice, CF altered the gut microbiota: it increased beneficial Actinobacteria (including Bifidobacterium and Blautia) and decreased Proteobacteria (including Desulfovibrio) (p < 0.05). Functional analysis showed that the HFD mice microbiota was enriched in genes linked to disease (e.g., lipid metabolism disorders, cancer, antibiotic resistance), whereas CF-enriched microbiota had genes for energy, carbohydrate, and pyruvate metabolism. Compared to microcrystalline cellulose, CF and MCC both alleviated HFD-induced obesity. In summary, cassava fiber helped prevent obesity in mice by modulating gut microbes, strengthening the gut barrier, and improving host metabolic balance. Full article
(This article belongs to the Special Issue Functional Foods, Gut Microbiota, and Health Benefits)
Show Figures

Graphical abstract

17 pages, 1989 KB  
Article
Optimal Dietary α-Starch Requirement and Its Effects on Growth and Metabolic Regulation in Chinese Hook Snout Carp (Opsariichthys bidens)
by Wenjing Cai, Xiaonian Luo, Jiao Li, Youjian Duan, Yong Wei, Yuxin Xing, Zongyun Hu and Chunyue Zhu
Biology 2025, 14(12), 1687; https://doi.org/10.3390/biology14121687 - 26 Nov 2025
Viewed by 457
Abstract
This study investigated the effects of dietary carbohydrate levels on growth performance, body composition, and hepatic expression of metabolic genes in Chinese hook snout carp (Opsariichthys bidens). Fish were fed five isonitrogenous diets with graded α-starch levels (8%, 14%, 20%, 26%, [...] Read more.
This study investigated the effects of dietary carbohydrate levels on growth performance, body composition, and hepatic expression of metabolic genes in Chinese hook snout carp (Opsariichthys bidens). Fish were fed five isonitrogenous diets with graded α-starch levels (8%, 14%, 20%, 26%, and 32%) for 56 days. The diet containing 14% α-starch significantly increased the weight gain rate (WGR) and specific growth rate (SGR) of O. bidens (p < 0.05). Both broken-line and polynomial regression analyses on WGR and SGR consistently indicated an optimal dietary α-starch level of approximately 14–17%. High carbohydrate diets significantly elevated plasma glucose, triglyceride, and cholesterol levels, as well as hepatosomatic and intraperitoneal fat indices. Gene expression analysis revealed that moderate carbohydrate intake upregulated lipoprotein lipase (lpl), hormone-sensitive lipase (hsl), and carnitine palmitoyltransferase 1 (cpt1) gene expressions, enhancing lipolysis and β-oxidation, whereas excessive carbohydrate intake (>26% α-starch) suppressed these pathways but strongly induced acc1 gene expressions, promoting lipogenesis. Additionally, glycogen metabolism genes (glycogen synthase (gys) and glycogen phosphorylase (pyg) and glycolysis-related phosphofructokinase (pfk) were responsive to carbohydrate supply, while oxidative metabolism gene cs was downregulated under excessive carbohydrate, implying reduced mitochondrial oxidative metabolism. Overall, O. bidens exhibited limited carbohydrate utilization, with optimal intake supporting growth and metabolic balance, whereas excessive intake redirected glucose toward glycogen and lipid accumulation, leading to metabolic imbalance. Full article
Show Figures

Figure 1

24 pages, 2429 KB  
Article
Protective Role of Ginsenoside F1-Enriched Extract (SGB121) in Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD)
by Bo Yoon Chang, In Kim, Hyungmin Park, Sunchang Kim and Sung Yeon Kim
Nutrients 2025, 17(23), 3693; https://doi.org/10.3390/nu17233693 - 25 Nov 2025
Viewed by 708
Abstract
Introduction/Objectives: Ginsenoside F1, a pharmacologically active saponin derived from Panax ginseng, exhibits diverse bioactivities, but its use is limited because it is difficult to purify and has high production costs. To overcome these challenges, a ginsenoside F1-enriched extract named SGB121 was developed. [...] Read more.
Introduction/Objectives: Ginsenoside F1, a pharmacologically active saponin derived from Panax ginseng, exhibits diverse bioactivities, but its use is limited because it is difficult to purify and has high production costs. To overcome these challenges, a ginsenoside F1-enriched extract named SGB121 was developed. This study aimed to evaluate the therapeutic efficacy of SGB121 in a high-fat, high-carbohydrate (HFHC) diet-induced metabolic dysfunction-associated fatty liver disease (MAFLD) mouse model and to elucidate its mechanism of action using F1-based cellular assays. Methods: Male C57BL/6 mice (6 weeks old) were fed an HFHC diet to induce MAFLD and were treated with SGB121. Hepatic lipid accumulation, oxidative stress markers, and metabolic parameters were analyzed. In parallel, human hepatocellular carcinoma (HepG2) cells exposed to free fatty acids (FFAs) were used to assess oxidative stress and lipid accumulation. Mechanistic studies were conducted using purified F1 to examine adenosine monophosphate-activated protein kinase (AMPK) activation and related pathways. Results: SGB121 reduced hepatic lipid accumulation, malondialdehyde (MDA) levels, and fasting insulin while restoring glutathione (GSH) content and improving the homeostasis model assessment of insulin resistance (HOMA-IR) in MAFLD mice. In FFA-treated HepG2 cells, both SGB121 and F1 decreased reactive oxygen species (ROS), suppressed sterol regulatory element-binding protein 1 (SREBP1), enhanced peroxisome proliferator-activated receptor-α (PPARα) and β-oxidation, and restored insulin receptor substrate (IRS)/protein kinase B (Akt)/glucose transporter 2 (GLUT2) signaling. Conclusions: SGB121 ameliorates MAFLD and related metabolic dysfunction through antioxidant, lipid-regulating, and insulin-sensitizing actions, highlighting its potential as a safe multifunctional nutraceutical for MAFLD management. Full article
Show Figures

Graphical abstract

21 pages, 915 KB  
Review
Dietary Modulation of the Gut Microbiota in Dogs and Cats and Its Role in Disease Management
by Benlu Yang, Shengwei Zhong, Jue Wang and Wanting Yu
Microorganisms 2025, 13(12), 2669; https://doi.org/10.3390/microorganisms13122669 - 24 Nov 2025
Viewed by 2309
Abstract
Food has a massive influence on the gut microbiota and is one of the most useful therapeutic levers in disease. Recent developments have highlighted how macronutrient balance, food format, and functional ingredients can regulate microbial diversity, metabolism, and host physiology in companion animals [...] Read more.
Food has a massive influence on the gut microbiota and is one of the most useful therapeutic levers in disease. Recent developments have highlighted how macronutrient balance, food format, and functional ingredients can regulate microbial diversity, metabolism, and host physiology in companion animals such as dogs and cats. This narrative review condenses evidence on the bidirectional gut microbiota–diet connection and on nutritional therapy for gastrointestinal, metabolic, renal, hepatic, and immune-mediated disorders. Protein-based diets including high or hydrolyzed protein, omega-3 acids, fermentative fiber, and probiotics can positively affect microbial composition, stimulate short-chain fatty acid synthesis, and enhance intestinal barrier functions. Conversely, excess fats or refined carbohydrates may cause dysbiosis, inflammation, and metabolic imbalances. Numerous studies have shown that therapeutic nutrition—e.g., low-protein renoprotective, hepatoprotective antioxidants, and allergen-elimination diets—holds enormous potential for treatment. In addition, fecal microbiota transplantation (FMT) can be used as an additive therapy for resistant gastrointestinal illnesses. Despite these developments, constraints remain in terms of standardization, study duration, and species-specific data, especially for cats. This review underscores dietary modification as a clinically actionable tool for microbiota-targeted therapy and calls for integrative, multi-omics research to translate microbiome modulation into precision nutrition for companion animals. Full article
(This article belongs to the Special Issue Dietary and Animal Gut Microbiota)
Show Figures

Figure 1

23 pages, 6248 KB  
Article
Radix Rehmanniae Praeparata Extract Enhances Liver Regeneration Through AMPK-Driven Metabolic Reprogramming
by Changmeng Li, Qi Zheng, Fanghong Li, Yinhao Zhang, Shuwen Duan, Jia Liu, Qi Han and Runping Liu
Nutrients 2025, 17(22), 3579; https://doi.org/10.3390/nu17223579 - 15 Nov 2025
Viewed by 2421
Abstract
Background: Liver regeneration is essential for restoring hepatic mass after injury or resection, with metabolic reprogramming as a critical driver. Radix Rehmanniae Praeparata (RRP), a traditional Chinese medicine for chronic liver diseases, regulates glucose and lipid metabolism. This study evaluated the effects of [...] Read more.
Background: Liver regeneration is essential for restoring hepatic mass after injury or resection, with metabolic reprogramming as a critical driver. Radix Rehmanniae Praeparata (RRP), a traditional Chinese medicine for chronic liver diseases, regulates glucose and lipid metabolism. This study evaluated the effects of RRP on liver regeneration and explored the underlying mechanisms. Methods: A 70% partial hepatectomy (PHx) mouse model was employed, and integrated transcriptomic and metabolomic analyses were conducted to characterize the global features of RRP-induced metabolic reprogramming and its association with hepatocyte proliferation. To further validate these findings, the AML12 hepatocyte cell line and primary mouse hepatocytes were used to identify key targets of RRP. Results: RRP significantly enhanced liver regeneration, as evidenced by the upregulation of hepatocyte proliferation markers. Transcriptomic, metabolomic, and biochemical analyses showed that RRP promoted lipid catabolism and H3K27ac remodeling-dependent hepatocyte proliferation by increasing acetyl-CoA flux. RRP also enhanced carbohydrate consumption and pentose phosphate pathway, as well as protecting mitochondrial integrity, which contribute to both energy production and nucleotide synthesis during cell cycle progression. Notably, RRP-induced AMPK activation was involved in these metabolic reprogramming events, since pharmacological inhibition of AMPK with Compound C attenuated the promotive effects of RRP on liver regeneration. Conclusions: RRP promotes liver regeneration by enhancing metabolic reprogramming mediated by AMPK activation, highlighting its therapeutic potential for metabolic adaptation and postoperative recovery in compromised liver. Full article
(This article belongs to the Section Phytochemicals and Human Health)
Show Figures

Figure 1

37 pages, 690 KB  
Article
Adaptive Ketogenic–Mediterranean Protocol (AKMP) in Real Clinical Practice: 14-Week Pre–Post Cohort Study on Glucolipid Markers and Safety
by Cayetano García-Gorrita, Nadia San Onofre, Juan F. Merino-Torres and Jose M. Soriano
Nutrients 2025, 17(22), 3559; https://doi.org/10.3390/nu17223559 - 14 Nov 2025
Viewed by 1380
Abstract
Background/Objectives: Overweight and obesity are associated with insulin resistance, atherogenic dyslipidemia, and low-grade inflammation. We evaluated analytical safety and within-person metabolic changes under the Adaptive Ketogenic–Mediterranean Protocol (AKMP) in real-world practice. Methods: Single arm, prospective pre–post cohort. We enrolled 112 adults; 105 completed [...] Read more.
Background/Objectives: Overweight and obesity are associated with insulin resistance, atherogenic dyslipidemia, and low-grade inflammation. We evaluated analytical safety and within-person metabolic changes under the Adaptive Ketogenic–Mediterranean Protocol (AKMP) in real-world practice. Methods: Single arm, prospective pre–post cohort. We enrolled 112 adults; 105 completed 14 weeks of AKMP (12 in nutritional ketosis ≤ 20 g carbohydrate/day + 2 of gradual reintroduction). Fasting venous samples were analyzed in accredited laboratories (glycolipid profile, hepatic–renal function, inflammatory markers; insulin, thyroid hormones, cortisol). HOMA-IR, TyG, and remnant cholesterol (RC) were calculated; body composition was measured by segmental bioimpedance. Paired analyses were used, with hierarchical gatekeeping for the conditional co-primary outcome and prespecified Δ~Δ correlations. Results: HOMA-IR −52.8% (Δ −1.80; p < 0.001) and RC −35.1% (Δ −10.64 mg/dL; p < 0.001); fasting glucose −13.7 mg/dL, insulin −5.9 μU/L; TyG −0.23 and TG/HDL-c −1.21 (all p < 0.001). Lipids: TG −35.1% and LDL-c −11.2%; HDL-c remained stable. Anthropometry: weight −14.85 kg (−14.7%) and trunk fat −4.88 kg (−22.2%) (p < 0.001). Safety: no serious adverse events; GGT −47.0%, eGFR +11.0%, and CRP −24.6% (p < 0.001). Prespecified correlations supported the internal consistency of the glycolipid axis (e.g., ΔHOMA-IR~ΔTyG; ΔRC~ΔHOMA-IR). Conclusions: In adults with overweight or obesity, the AKMP was associated with improvements in the glucose–insulin axis, atherogenic profile (RC, TG/HDL-c, TG), and body composition, while maintaining a favorable safety profile. The protocol appears feasible in clinical practice and monitorable with routine laboratory tests, although randomized controlled trials are needed to confirm causality and long-term sustainability. Full article
(This article belongs to the Section Clinical Nutrition)
Show Figures

Figure 1

22 pages, 3492 KB  
Article
Minimum Dietary Fat Threshold for Effective Ketogenesis and Obesity Control in Mice
by Jiawen Shou, Xingchen Dong, Fei Sun, Jia Li, Huiren Wang, Qing Ai, Michael Pellizzon and Ting Fu
Nutrients 2025, 17(20), 3203; https://doi.org/10.3390/nu17203203 - 12 Oct 2025
Viewed by 1291
Abstract
Background/Objectives: Ketogenic diets (KDs), defined by very low carbohydrate and high fat content, are widely studied for obesity and metabolic disease. However, KD formulations vary from 60–95% fat, leading to inconsistent induction of ketogenesis and variable outcomes. The fat threshold required for [...] Read more.
Background/Objectives: Ketogenic diets (KDs), defined by very low carbohydrate and high fat content, are widely studied for obesity and metabolic disease. However, KD formulations vary from 60–95% fat, leading to inconsistent induction of ketogenesis and variable outcomes. The fat threshold required for sustained ketosis, and the tissue-specific programs that mediate KD efficacy, remain unclear. Methods: We evaluated multiple KD formulations (80–95% fat) in C57BL/6J wild-type (WT) and diet-induced obese (DIO) mice. Plasma, hepatic, and intestinal β-hydroxybutyrate (BHB) were measured together with expression of ketogenesis and fatty acid oxidation genes. Body weight, adipose distribution, and liver morphology were assessed under both direct feeding and therapeutic settings. Results: In WT mice, only diets exceeding 85% fat induced robust ketogenesis, reflected by elevated BHB and hepatic upregulation of Cd36, Cpt1a, Acat1, and Hmgcs2. Moderate KDs (80–85%) failed to trigger ketosis and resembled high-fat feeding. In obese mice, an 80% KD lowered fasting glucose without reducing body weight, whereas a 90% KD promoted systemic ketosis, weight loss, and adipose reduction. Interestingly, hepatic transcriptional programs for fatty acid oxidation and ketogenesis were suppressed under 90% KD despite elevated BHB, suggesting reliance on substrate availability and peripheral utilization. In contrast, intestinal Hmgcs2 was strongly induced in both WT and DIO mice, with Oxct1 upregulated only in obesity, indicating local ketone production and consumption. Conclusions: These findings identify > 85% dietary fat as a threshold for sustained ketosis and highlight distinct liver–intestine contributions, underscoring ketogenesis as the central driver of KD’s anti-obesity benefits. Full article
(This article belongs to the Section Nutrition and Public Health)
Show Figures

Figure 1

49 pages, 10722 KB  
Review
Triglycerides, Glucose Metabolism, and Type 2 Diabetes
by Yutang Wang
Int. J. Mol. Sci. 2025, 26(20), 9910; https://doi.org/10.3390/ijms26209910 - 11 Oct 2025
Cited by 3 | Viewed by 5954
Abstract
Type 2 diabetes is a major global health burden, causing approximately 2 million deaths annually. Recent studies have revealed a strong positive correlation between elevated triglyceride levels and plasma glucose, as well as increased prevalence, incidence, and mortality of type 2 diabetes, suggesting [...] Read more.
Type 2 diabetes is a major global health burden, causing approximately 2 million deaths annually. Recent studies have revealed a strong positive correlation between elevated triglyceride levels and plasma glucose, as well as increased prevalence, incidence, and mortality of type 2 diabetes, suggesting a potential causal link. This review explores the metabolic interconversion between triglycerides and glucose, emphasizing how excess carbohydrate intake leads to ectopic triglyceride accumulation, which in turn enhances hepatic gluconeogenesis. It highlights key signaling pathways through which ectopic triglyceride deposition drives insulin resistance, hyperinsulinemia, β-cell dysfunction and apoptosis, and increased glucose production—central mechanisms in diabetes pathogenesis. Evidence from clinical interventions, such as the reversal of type 2 diabetes through bariatric surgery and dietary energy restriction, supports the hypothesis that ectopic triglyceride accumulation is a driving factor. Furthermore, this review explains why omega-3 fatty acids and niacin, in contrast to fibrates, do not protect against type 2 diabetes, despite lowering triglycerides. Overall, this review emphasizes the contribution of ectopic triglyceride accumulation—driven by obesity, hypertriglyceridemia, excessive consumption of carbohydrates and fats, and physical inactivity—to the onset and progression of type 2 diabetes, offering valuable insights into potential therapeutic strategies. Full article
(This article belongs to the Special Issue The Role of Lipids in Health and Diseases)
Show Figures

Figure 1

24 pages, 6712 KB  
Article
Biomarkers Characterizing the Onset of Dietary-Induced Hepatocellular Injury and Visceral Obesity in a Rat Experimental Model: Possible Anti-Inflammatory Effects of Steviol Glycosides
by Krastina Trifonova, Penka Yonkova and Petko Dzhelebov
Metabolites 2025, 15(10), 656; https://doi.org/10.3390/metabo15100656 - 4 Oct 2025
Viewed by 992
Abstract
Background: The aim of the present study is to compare the potential of a high-fat diet, a high-carbohydrate diet, and a high-fat, high-carbohydrate diet to induce liver injury and visceral obesity within a period of five weeks, identify the pattern and degree of [...] Read more.
Background: The aim of the present study is to compare the potential of a high-fat diet, a high-carbohydrate diet, and a high-fat, high-carbohydrate diet to induce liver injury and visceral obesity within a period of five weeks, identify the pattern and degree of hepatic changes at the tissue level, identify the earliest metabolic markers of specific liver changes induced by each type of diet, and to test the possible beneficial effects of steviol glycosides in a rat experimental model. Methods: Wistar rats (n = 56) were divided into seven groups as follows: group BD (before diet), group SD (standard diet), group HFD (high-fat diet), group HCHD (high-carbohydrate diet), group HFHCHD (high-fat high-carbohydrate diet), group SDS (standard diet supplemented with Stevia extract), and group HFDS (high-fat diet supplemented with Stevia extract). Results: Total cholesterol concentrations (2.02 ± 0.22 mmol/L) increased in the HFD group (2.56 ± 0.82 mmol/L) and in the HFDS group (2.89 ± 0.48 mmol/L). The VLDL values before diets were 0.27 ± 0.11 mmol/L and increased most significantly in the HFHCHD group—1.14 ± 0.62 mmol/L. The baseline ALT values (88.4 ± 10.6 U/L) increased in the HFD group (128.13 ± 19.5 U/L) and the HFDS group (127.00 ± 17.74 U/L). Similar increases were registered in the AST/ALT ratio and ALP. Total bilirubin (7.10 ± 1.39 μmol/L) increased in HFD group (27.86 ± 17.01 μmol/L). Serum NO had the lowest values in groups fed diets supplemented with steviol glycosides. All high-calorie diets induced hepatocellular injury. The mass of the perirenal fat depot and cross-sectional area of adipocytes were highest in HFD, HFHCHD, and HFDS groups. Conclusion: High-calorie diets have the potential to induce visceral obesity and hepatocellular injury within a very short period of time, which produces characteristic histological changes and specific biochemical profile. Steviol glycosides may alleviate some aspects of the inflammatory response, but findings about lipid profile parameters and liver enzymes are controversial. Full article
(This article belongs to the Special Issue Metabolic Changes in Diet-Mediated Inflammatory Diseases)
Show Figures

Figure 1

13 pages, 670 KB  
Review
Characteristic Gene Alterations During Fatty Acid Metabolism in the Goose Liver
by Anna Koseniuk
Genes 2025, 16(10), 1137; https://doi.org/10.3390/genes16101137 - 25 Sep 2025
Viewed by 1230
Abstract
The development of hepatic steatosis in geese is a complex, multistage process involving genes related to lipid synthesis, transport, storage, and metabolism. Key genes activated during this process include ME1 (malic enzyme 1), SCD1 (stearoyl-CoA desaturase), ACSL1 (acyl-CoA synthetase long-chain family member 1), [...] Read more.
The development of hepatic steatosis in geese is a complex, multistage process involving genes related to lipid synthesis, transport, storage, and metabolism. Key genes activated during this process include ME1 (malic enzyme 1), SCD1 (stearoyl-CoA desaturase), ACSL1 (acyl-CoA synthetase long-chain family member 1), and ELOVL6 (elongation of very-long-chain fatty acids protein 6). The expression of these genes varies depending on the tissue, breed, and metabolic context. Geese possess a unique ability to develop hepatic steatosis (fatty liver) without accompanying inflammation or liver damage. This condition typically arises from overfeeding, either through carbohydrates or fats, leading to significant triglyceride accumulation in hepatocytes. Importantly, this state remains reversible and is considered non-pathological. The physiological and molecular changes observed in overfed geese, particularly regarding liver lipid accumulation and serum enzyme activity, closely resemble those found in human non-alcoholic fatty liver disease (NAFLD). This similarity makes geese an excellent biomedical model for studying NAFLD. Overfeeding initiates a cascade of enzymatic reactions that regulate lipid metabolism at the genetic level. These reactions decrease circulating free fatty acids and glucose while promoting triglyceride storage in the liver. The aim of this study is to synthesize current knowledge on the genetic regulation of fatty acid metabolism in geese, highlighting how these genes coordinate the processes of activation, desaturation, synthesis, and elongation during induced steatosis. Moreover, the summarized effects of different diet supplements will enhance goose feeding strategies for foie gras production. Full article
(This article belongs to the Special Issue Genetic Breeding of Poultry)
Show Figures

Figure 1

22 pages, 2710 KB  
Article
Divergent Hepatic Outcomes of Chronic Ketone Supplementation: Ketone Salts Preserve Liver Health While Ketone Esters and Precursors Drive Inflammation and Steatosis
by Csilla Ari and Dominic P. D’Agostino
Pharmaceuticals 2025, 18(10), 1436; https://doi.org/10.3390/ph18101436 - 25 Sep 2025
Cited by 1 | Viewed by 7095
Abstract
Background/Objectives: Exogenous ketone supplements elevate circulating ketones without carbohydrate restriction, but their long-term hepatic safety remains unclear. This study evaluated the formulation-dependent impact of chronic ketone supplementation on liver histopathology, inflammatory signaling, and systemic biomarkers in rats. Methods: Male Sprague-Dawley rats were orally [...] Read more.
Background/Objectives: Exogenous ketone supplements elevate circulating ketones without carbohydrate restriction, but their long-term hepatic safety remains unclear. This study evaluated the formulation-dependent impact of chronic ketone supplementation on liver histopathology, inflammatory signaling, and systemic biomarkers in rats. Methods: Male Sprague-Dawley rats were orally administered 1,3-butanediol (BD), medium-chain triglycerides (MCTs), ketone ester (KE), ketone electrolytes/salts (KSs), or a ketone salt–MCT combination (KSMCT) for 4 weeks. In a separate arm, animals received standard diet (SD), or SD supplemented with low-dose KE (LKE) or high-dose KE (HKE), for 83 days. Liver structure was assessed by hematoxylin and eosin staining with quantification of red blood cell density and lipid accumulation. Inflammatory and metabolic responses were evaluated by TNF-α and arginase immunohistochemistry. Serum biochemistry included glucose, proteins, electrolytes, and liver and kidney function markers. Results: BD and KE induced macrovesicular steatosis, vascular congestion, and elevated TNF-α and arginase expression, consistent with hepatic stress. MCT caused moderate hepatocellular ballooning and lipid deposition, whereas KS preserved near-normal hepatic morphology. KSMCT produced intermediate effects, reducing lipid accumulation and TNF-α compared with MCT or KE alone. KE supplementation caused dose-dependent reductions in globulin and elevations in creatinine, while HKE reduced sodium and glucose levels. Conclusions: Chronic hepatic responses to exogenous ketones are highly formulation dependent. KS demonstrated the most favorable safety profile under the tested conditions, maintaining normal hepatic structure, while BD and KE elicited adverse changes. Formulation choice is critical for the safe long-term use of exogenous ketones. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Graphical abstract

35 pages, 3238 KB  
Review
The Ketogenic Diet Through a Metabolomic Lens: Biochemical Pathways, Therapeutic Applications, and Analytical Challenges
by Katarzyna Idzikowska, Paulina Gątarek, Anna Gajda, Piotr Safiński, Lukasz Przyslo and Joanna Kałużna-Czaplińska
Nutrients 2025, 17(18), 2969; https://doi.org/10.3390/nu17182969 - 16 Sep 2025
Cited by 3 | Viewed by 4198
Abstract
Background: The ketogenic diet (KD), a high-fat and low-carbohydrate dietary approach, has been used therapeutically in drug-resistant epilepsy and other neurological and metabolic disorders. Recent interest has shifted toward understanding its broader metabolic effects through metabolomics. This review aims to summarize current knowledge [...] Read more.
Background: The ketogenic diet (KD), a high-fat and low-carbohydrate dietary approach, has been used therapeutically in drug-resistant epilepsy and other neurological and metabolic disorders. Recent interest has shifted toward understanding its broader metabolic effects through metabolomics. This review aims to summarize current knowledge on the biochemical mechanisms and therapeutic implications of the KD, with a particular focus on metabolomic profiling and neurological health. Methods: This narrative review synthesizes findings from the last five years of metabolomic studies investigating the biochemical consequences of the KD and its variants, including the classical KD, modified Atkins diet (MAD), medium-chain triglyceride diet (MCT), and low glycemic index treatment (LGIT). The review integrates data on analytical techniques, such as liquid chromatography–mass spectrometry (LC-MS) and gas chromatography–mass spectrometry (GC-MS), and evaluates alterations in key metabolic pathways. Results: The KD significantly modulates energy metabolism, shifting adenosine triphosphate (ATP) production from glycolysis to fatty acid oxidation and ketone body utilization. It affects mitochondrial function, one-carbon metabolism, redox balance, neurotransmitter regulation, and gut–brain axis signaling. Metabolomic profiling has identified β-hydroxybutyrate (βHB) as a key regulatory metabolite influencing mitochondrial respiration. Long-term KD use may impact renal and hepatic function, necessitating clinical caution and individualized nutritional monitoring. Conclusions: Metabolomic analysis provides critical insights into the multifaceted effects of the KD, supporting its role as a targeted metabolic therapy in neurological diseases. However, potential risks linked to prolonged ketosis warrant further investigation. Future studies should focus on personalized applications and long-term safety profiles of KD variants across patient populations. Full article
(This article belongs to the Special Issue Neurological Disorders: Diets and Nutrition)
Show Figures

Figure 1

Back to TopTop