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

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
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,336)

Search Parameters:
Keywords = SCFAs (Short-chain fatty acids)

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 5256 KiB  
Article
Impact of Alginate Oligosaccharides on Ovarian Performance and the Gut Microbial Community in Mice with D-Galactose-Induced Premature Ovarian Insufficiency
by Yan Zhang, Hongda Pan, Dao Xiang, Hexuan Qu and Shuang Liang
Antioxidants 2025, 14(8), 962; https://doi.org/10.3390/antiox14080962 - 5 Aug 2025
Abstract
Premature ovarian insufficiency (POI) is an important factor in female infertility and is often associated with oxidative stress. Alginate oligosaccharides (AOSs), derived from the degradation of alginate, have been demonstrated to have protective effects against various oxidative stress-related diseases. However, the impact of [...] Read more.
Premature ovarian insufficiency (POI) is an important factor in female infertility and is often associated with oxidative stress. Alginate oligosaccharides (AOSs), derived from the degradation of alginate, have been demonstrated to have protective effects against various oxidative stress-related diseases. However, the impact of AOSs on POI has not been previously explored. The current study explored the effects of AOSs on ovarian dysfunction in a mouse model of POI induced by D-galactose (D-gal). Female C57BL/6 mice were randomly divided into five groups: the control (CON), POI model (D-gal), and low-, medium-, and high-dose AOS groups (AOS-L, 100 mg/kg/day; AOS-M, 150 mg/kg/day; AOS-H, 200 mg/kg/day). For 42 consecutive days, mice in the D-gal, AOS-L, AOS-M, and AOS-H groups received daily intraperitoneal injections of D-gal (200 mg/kg/day), whereas those in the CON group received equivalent volumes of sterile saline. Following D-gal injection, AOSs were administered via gavage at the specified doses; mice in the CON and D-gal groups received sterile saline instead. AOS treatment markedly improved estrous cycle irregularities, normalized serum hormone levels, reduced granulosa cell apoptosis, and increased follicle counts in POI mice. Moreover, AOSs significantly reduced ovarian oxidative stress and senescence in POI mice, as indicated by lower levels of malondialdehyde (MDA), higher activities of catalase (CAT) and superoxide dismutase (SOD), and decreased protein expression of 4-hydroxynonenal (4-HNE), nitrotyrosine (NTY), 8-hydroxydeoxyguanosine (8-OHdG), and p16 in ovarian tissue. Analysis of the gut microbiota through 16S rRNA gene sequencing and short-chain fatty acid (SCFA) analysis revealed significant differences in gut microbiota composition and SCFA levels (acetic acid and total SCFAs) between control and D-gal-induced POI mice. These differences were largely alleviated by AOS treatment. AOSs changed the gut microbiota by increasing the abundance of Ligilactobacillus and decreasing the abundance of Clostridiales, Clostridiaceae, Marinifilaceae, and Clostridium_T. Additionally, AOSs mitigated the decline in acetic acid and total SCFA levels observed in POI mice. Notably, the total SCFA level was significantly correlated with the abundance of Ligilactobacillus, Marinifilaceae, and Clostridium_T. In conclusion, AOS intervention effectively mitigates ovarian oxidative stress, restores gut microbiota homeostasis, and regulates the microbiota–SCFA axis, collectively improving D-gal-induced POI. Therefore, AOSs represent a promising therapeutic strategy for POI management. Full article
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)
Show Figures

Figure 1

18 pages, 3360 KiB  
Article
Hydrogen Sulfide Has a Minor Impact on Human Gut Microbiota Across Age Groups
by Linshu Liu, Johanna M. S. Lemons, Jenni Firrman, Karley K. Mahalak, Venkateswari J. Chetty, Adrienne B. Narrowe, Stephanie Higgins, Ahmed M. Moustafa, Aurélien Baudot, Stef Deyaert and Pieter Van den Abbeele
Sci 2025, 7(3), 102; https://doi.org/10.3390/sci7030102 - 1 Aug 2025
Viewed by 128
Abstract
Hydrogen sulfide (H2S) can be produced from the metabolism of foods containing sulfur in the gastrointestinal tract (GIT). At low doses, H2S regulates the gut microbial community and supports GIT health, but depending on dose, age, and individual health [...] Read more.
Hydrogen sulfide (H2S) can be produced from the metabolism of foods containing sulfur in the gastrointestinal tract (GIT). At low doses, H2S regulates the gut microbial community and supports GIT health, but depending on dose, age, and individual health conditions, it may also contribute to inflammatory responses and gut barrier dysfunction. Controlling H2S production in the GIT is important for maintaining a healthy gut microbiome. However, research on this subject is limited due to the gaseous nature of the chemical and the difficulty of accessing the GIT in situ. In the present ex vivo experiment, we used a single-dose sodium sulfide preparation (SSP) as a H2S precursor to test the effect of H2S on the human gut microbiome across different age groups, including breastfed infants, toddlers, adults, and older adults. Metagenomic sequencing and metabolite measurements revealed that the development of the gut microbial community and the production of short-chain fatty-acids (SCFAs) were age-dependent; that the infant and the older adult groups were more sensitive to SSP exposure; that exogeneous SSP suppressed SCFA production across all age groups, except for butyrate in the older adult group, suggesting that H2S selectively favors specific gut microbial processes. Full article
(This article belongs to the Section Biology Research and Life Sciences)
Show Figures

Figure 1

23 pages, 766 KiB  
Review
Pathophysiological Links Between Inflammatory Bowel Disease and Cardiovascular Disease: The Role of Dysbiosis and Emerging Biomarkers
by Roko Šantić, Nikola Pavlović, Marko Kumrić, Marino Vilović and Joško Božić
Biomedicines 2025, 13(8), 1864; https://doi.org/10.3390/biomedicines13081864 - 31 Jul 2025
Viewed by 148
Abstract
This review introduces a novel integrative framework linking gut dysbiosis, systemic inflammation, and cardiovascular risk in patients with inflammatory bowel disease (IBD). We highlight emerging biomarkers, including short-chain fatty acids (SCFAs), calprotectin, and zonulin, that reflect alterations in the gut microbiome and increased [...] Read more.
This review introduces a novel integrative framework linking gut dysbiosis, systemic inflammation, and cardiovascular risk in patients with inflammatory bowel disease (IBD). We highlight emerging biomarkers, including short-chain fatty acids (SCFAs), calprotectin, and zonulin, that reflect alterations in the gut microbiome and increased intestinal permeability, which contribute to cardiovascular pathology. Cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality worldwide, and recent evidence identifies IBD, encompassing ulcerative colitis (UC) and Crohn’s disease (CD), as a significant non-traditional risk factor for CVD. This review synthesizes current knowledge on how dysbiosis-driven inflammation in IBD patients exacerbates endothelial dysfunction, hypercoagulability, and atherosclerosis, even in the absence of traditional risk factors. Additionally, we discuss how commonly used IBD therapies may modulate cardiovascular risk. Understanding these multifactorial mechanisms and validating reliable biomarkers are essential for improving cardiovascular risk stratification and guiding targeted prevention strategies in this vulnerable population. Full article
Show Figures

Figure 1

15 pages, 1360 KiB  
Systematic Review
Prebiotics Improve Blood Pressure Control by Modulating Gut Microbiome Composition and Function: A Systematic Review and Meta-Analysis
by Abdulwhab Shremo Msdi, Elisabeth M. Wang and Kevin W. Garey
Nutrients 2025, 17(15), 2502; https://doi.org/10.3390/nu17152502 - 30 Jul 2025
Viewed by 410
Abstract
Background: Ingestion of dietary fibers (DFs) is a safe and accessible intervention associated with reductions in blood pressure (BP) and cardiovascular mortality. However, the mechanisms underlying the antihypertensive effects of DFs remain poorly defined. This systematic review and meta-analysis evaluates how DFs influence [...] Read more.
Background: Ingestion of dietary fibers (DFs) is a safe and accessible intervention associated with reductions in blood pressure (BP) and cardiovascular mortality. However, the mechanisms underlying the antihypertensive effects of DFs remain poorly defined. This systematic review and meta-analysis evaluates how DFs influence BP regulation by modulating gut microbial composition and enhancing short-chain fatty acid (SCFA) production. Methods: MEDLINE and EMBASE were systematically searched for interventional studies published between January 2014 and December 2024. Eligible studies assessed the effects of DFs or other prebiotics on systolic BP (SBP) and diastolic BP (DBP) in addition to changes in gut microbial or SCFA composition. Results: Of the 3010 records screened, nineteen studies met the inclusion criteria (seven human, twelve animal). A random-effects meta-analysis was conducted on six human trials reporting post-intervention BP values. Prebiotics were the primary intervention. In hypertensive cohorts, prebiotics significantly reduced SBP (−8.5 mmHg; 95% CI: −13.9, −3.1) and DBP (−5.2 mmHg; 95% CI: −8.5, −2.0). A pooled analysis of hypertensive and non-hypertensive patients showed non-significant reductions in SBP (−4.5 mmHg; 95% CI: −9.3, 0.3) and DBP (−2.5 mmHg; 95% CI: −5.4, 0.4). Animal studies consistently showed BP-lowering effects across diverse etiologies. Prebiotic interventions restored bacterial genera known to metabolize DFs to SCFAs (e.g., Bifidobacteria, Akkermansia, and Coprococcus) and increased SCFA levels. Mechanistically, SCFAs act along gut–organ axes to modulate immune, vascular, and neurohormonal pathways involved in BP regulation. Conclusions: Prebiotic supplementation is a promising strategy to reestablish BP homeostasis in hypertensive patients. Benefits are likely mediated through modulation of the gut microbiota and enhanced SCFA production. Full article
(This article belongs to the Special Issue Probiotics and Prebiotics: Past, Present and Future)
Show Figures

Graphical abstract

25 pages, 1199 KiB  
Review
Gut-Microbiota-Derived Metabolites and Probiotic Strategies in Colorectal Cancer: Implications for Disease Modulation and Precision Therapy
by Yi-Chu Yang, Shih-Chang Chang, Chih-Sheng Hung, Ming-Hung Shen, Ching-Long Lai and Chi-Jung Huang
Nutrients 2025, 17(15), 2501; https://doi.org/10.3390/nu17152501 - 30 Jul 2025
Viewed by 535
Abstract
The human gut microbiota significantly influences host health through its metabolic products and interaction with immune, neural, and metabolic systems. Among these, short-chain fatty acids (SCFAs), especially butyrate, play key roles in maintaining gut barrier integrity, modulating inflammation, and supporting metabolic regulation. Dysbiosis [...] Read more.
The human gut microbiota significantly influences host health through its metabolic products and interaction with immune, neural, and metabolic systems. Among these, short-chain fatty acids (SCFAs), especially butyrate, play key roles in maintaining gut barrier integrity, modulating inflammation, and supporting metabolic regulation. Dysbiosis is increasingly linked to diverse conditions such as gastrointestinal, metabolic, and neuropsychiatric disorders, cardiovascular diseases, and colorectal cancer (CRC). Probiotics offer therapeutic potential by restoring microbial balance, enhancing epithelial defenses, and modulating immune responses. This review highlights the physiological functions of gut microbiota and SCFAs, with a particular focus on butyrate’s anti-inflammatory and anti-cancer effects in CRC. It also examines emerging microbial therapies like probiotics, synbiotics, postbiotics, and engineered microbes. Emphasis is placed on the need for precision microbiome medicine, tailored to individual host–microbiome interactions and metabolomic profiles. These insights underscore the promising role of gut microbiota modulation in advancing preventive and personalized healthcare. Full article
(This article belongs to the Special Issue Diet, Gut Microbiota, and Gastrointestinal Disease)
Show Figures

Graphical abstract

28 pages, 1184 KiB  
Review
Immune Modulation by Microbiota and Its Possible Impact on Polyomavirus Infection
by Giorgia Cianci, Gloria Maini, Matteo Ferraresi, Giulia Pezzi, Daria Bortolotti, Sabrina Rizzo, Silvia Beltrami and Giovanna Schiuma
Pathogens 2025, 14(8), 747; https://doi.org/10.3390/pathogens14080747 - 30 Jul 2025
Viewed by 396
Abstract
Polyomaviruses are a family of small DNA viruses capable of establishing persistent infections, and they can pose significant pathogenic risks in immunocompromised hosts. While traditionally studied in the context of viral reactivation and immune suppression, recent evidence has highlighted the gut microbiota as [...] Read more.
Polyomaviruses are a family of small DNA viruses capable of establishing persistent infections, and they can pose significant pathogenic risks in immunocompromised hosts. While traditionally studied in the context of viral reactivation and immune suppression, recent evidence has highlighted the gut microbiota as a critical regulator of host immunity and viral pathogenesis. This review examines the complex interactions between polyomaviruses, the immune system, and intestinal microbiota, emphasizing the role of short-chain fatty acids (SCFAs) in modulating antiviral responses. We explore how dysbiosis may facilitate viral replication, reactivation, and immune escape and also consider how polyomavirus infection can, in turn, alter microbial composition. Particular attention is given to the Firmicutes/Bacteroidetes ratio as a potential biomarker of infection risk and immune status. Therapeutic strategies targeting the microbiota, including prebiotics, probiotics, and fecal microbiota transplantation (FMT), are discussed as innovative adjuncts to immune-based therapies. Understanding these tri-directional interactions may offer new avenues for mitigating disease severity and improving patient outcomes during viral reactivation. Full article
Show Figures

Figure 1

22 pages, 4967 KiB  
Article
Therapeutic Potential of Kelp Fucoidan in Rebiosis of Gut Microflora and Immune Homeostasis in Cyclophosphamide-Induced Immunosuppressed Mice
by Yaqing Liu, Ruining Kang, Yanfei Zhao, Heng Zhang, Qingfeng Rong, Shaoxuan Yu, Yaoguang Chang, Zhengpeng Wei and Lanlan Zhu
Foods 2025, 14(15), 2662; https://doi.org/10.3390/foods14152662 - 29 Jul 2025
Viewed by 266
Abstract
Recent studies indicate that fucoidan may play a crucial role in the metabolism and biological function of the intestinal flora. This study investigates the therapeutic potential of kelp fucoidan on the gut microbiota and immune homeostasis of cyclophosphamide-induced immunosuppressed mice. An immunosuppressive mouse [...] Read more.
Recent studies indicate that fucoidan may play a crucial role in the metabolism and biological function of the intestinal flora. This study investigates the therapeutic potential of kelp fucoidan on the gut microbiota and immune homeostasis of cyclophosphamide-induced immunosuppressed mice. An immunosuppressive mouse model was established using cyclophosphamide, followed by administration of various kelp fucoidan doses (low-dose fucoidan: 50 mg/(kg·bw)/d, medium-dose fucoidan: 100 mg/(kg·bw)/d, and high-dose fucoidan: 150 mg/(kg·bw)/d) to the experimental groups. Changes in the gut microbiota structure were analyzed using 16S rRNA high-throughput sequencing, alongside simultaneous measurement of serum immune indicators and levels of short-chain fatty acids (SCFAs). Results indicate that kelp fucoidan significantly improved the thymus and spleen indices in immunosuppressed mice (p < 0.05) and elevated serum levels of IgM, IgG and IL-4. Post-kelp fucoidan intervention, there was significant alteration in microbiota ecosystem restructuring, such as proliferation in probiotics, including Lactobacillus and Bifidobacterium, while opportunistic pathogens, such as Enterococcus and Escherichia coli, decreased. Furthermore, the levels of acetic, propionic, and butyric acids in the colonic contents of the kelp fucoidan group significantly improved (p < 0.01). This research demonstrates that kelp fucoidan enhances immune function in immunosuppressed mice by modulating gut microbiota balance and promoting short-chain fatty acid production. Full article
(This article belongs to the Section Food Nutrition)
Show Figures

Figure 1

20 pages, 2441 KiB  
Article
Dysfunction and Metabolic Reprogramming of Gut Regulatory T Cells in HIV-Infected Immunological Non-Responders
by Minrui Yu, Mengmeng Qu, Zerui Wang, Cheng Zhen, Baopeng Yang, Yi Zhang, Huihuang Huang, Chao Zhang, Jinwen Song, Xing Fan, Ruonan Xu, Yan-Mei Jiao and Fu-Sheng Wang
Cells 2025, 14(15), 1164; https://doi.org/10.3390/cells14151164 - 29 Jul 2025
Viewed by 359
Abstract
Disruption of the gut microenvironment is a hallmark of HIV infection, where regulatory T cells (Tregs) play a critical role in maintaining gut homeostasis. However, the mechanisms by which gut Tregs contribute to immune reconstitution failure in HIV-infected individuals remain poorly understood. In [...] Read more.
Disruption of the gut microenvironment is a hallmark of HIV infection, where regulatory T cells (Tregs) play a critical role in maintaining gut homeostasis. However, the mechanisms by which gut Tregs contribute to immune reconstitution failure in HIV-infected individuals remain poorly understood. In this study, we employed single-cell RNA sequencing (scRNA-seq) to analyze gut Treg populations across three cohorts: eight immunological responders (IRs), three immunological non-responders (INRs), and four HIV-negative controls (NCs). Our findings revealed that INRs exhibit an increased proportion of gut Tregs but with significant functional impairments, including reduced suppressive capacity and heightened apoptotic activity. Notably, these Tregs underwent metabolic reprogramming in INRs, marked by an upregulation of glycolysis-related genes and a downregulation of the oxidative phosphorylation (OXPHOS) pathway. Additionally, both the abundance of short-chain fatty acid (SCFA)-producing bacteria and SCFA concentrations were reduced in INRs. In vitro SCFA supplementation restored Treg function by enhancing suppressive capacity, reducing early apoptosis, and rebalancing cellular energy metabolism from glycolysis to OXPHOS. These findings provide a comprehensive characterization of gut Treg dysfunction in INRs and underscore the therapeutic potential of targeting gut Tregs through microbiota and metabolite supplementation to improve immune reconstitution in HIV-infected individuals. Full article
(This article belongs to the Special Issue Immune Response in HIV Infection, Pathogenesis and Persistence)
Show Figures

Graphical abstract

17 pages, 2387 KiB  
Article
Application of Lactobacillus helveticus KLDS 1.1105 Postbiotics for Resisting Pathogenic Bacteria Infection in the Intestine
by Peng Du, Jiaying Liu, Chengwen Hu, Jianing Zhang, Miao Li, Yu Xin, Libo Liu, Aili Li and Chun Li
Foods 2025, 14(15), 2659; https://doi.org/10.3390/foods14152659 - 29 Jul 2025
Viewed by 311
Abstract
Postbiotics, defined as metabolites produced by probiotics, encompass both bacterial cells and their metabolic byproducts, and offer significant health benefits to the host. However, there are relatively few reports on their effects on intestinal microbiota. In this study, we investigated the components, total [...] Read more.
Postbiotics, defined as metabolites produced by probiotics, encompass both bacterial cells and their metabolic byproducts, and offer significant health benefits to the host. However, there are relatively few reports on their effects on intestinal microbiota. In this study, we investigated the components, total antioxidant capacity of Lactobacillus helveticus postbiotics (LHPs) and their impact on intestinal flora using the Simulator for Human Intestinal Microecology Simulation (SHIME). The results indicate that the primary components of postbiotics include polysaccharides, proteins, and organic acids. Furthermore, LHPs have a strong ability to inhibit the growth of harmful bacteria while promoting the growth of probiotics. Additionally, LHPs significantly increased the total antioxidant capacity in the intestine and regulated the balance of intestinal microbiota. Notably, there was also a significant increase in the content of short-chain fatty acids (SCFAs) in the intestine. Overall, LHPs have the potential to aid in the prevention and treatment of diseases by enhancing gut microbiology. Full article
Show Figures

Graphical abstract

21 pages, 2030 KiB  
Article
Restoring Balance: Probiotic Modulation of Microbiota, Metabolism, and Inflammation in SSRI-Induced Dysbiosis Using the SHIME® Model
by Marina Toscano de Oliveira, Fellipe Lopes de Oliveira, Mateus Kawata Salgaço, Victoria Mesa, Adilson Sartoratto, Kalil Duailibi, Breno Vilas Boas Raimundo, Williams Santos Ramos and Katia Sivieri
Pharmaceuticals 2025, 18(8), 1132; https://doi.org/10.3390/ph18081132 - 29 Jul 2025
Viewed by 553
Abstract
Background/Objectives: Selective serotonin reuptake inhibitors (SSRIs), widely prescribed for anxiety disorders, may negatively impact the gut microbiota, contributing to dysbiosis. Considering the gut–brain axis’s importance in mental health, probiotics could represent an effective adjunctive strategy. This study evaluated the effects of Lactobacillus helveticus [...] Read more.
Background/Objectives: Selective serotonin reuptake inhibitors (SSRIs), widely prescribed for anxiety disorders, may negatively impact the gut microbiota, contributing to dysbiosis. Considering the gut–brain axis’s importance in mental health, probiotics could represent an effective adjunctive strategy. This study evaluated the effects of Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 on microbiota composition, metabolic activity, and immune markers in fecal samples from patients with anxiety on SSRIs, using the SHIME® (Simulator of the Human Intestinal Microbial Ecosystem) model. Methods: The fecal microbiotas of four patients using sertraline or escitalopram were inoculated in SHIME® reactors simulating the ascending colon. After stabilization, a 14-day probiotic intervention was performed. Microbial composition was assessed by 16S rRNA sequencing. Short-chain fatty acids (SCFAs), ammonia, and GABA were measured, along with the prebiotic index (PI). Intestinal barrier integrity was evaluated via transepithelial electrical resistance (TEER), and cytokine levels (IL-6, IL-8, IL-10, TNF-α) were analyzed using a Caco-2/THP-1 co-culture system. The statistical design employed in this study for the analysis of prebiotic index, metabolites, intestinal barrier integrity and cytokines levels was a repeated measures ANOVA, complemented by post hoc Tukey’s tests to assess differences across treatment groups. For the 16S rRNA sequencing data, alpha diversity was assessed using multiple metrics, including the Shannon, Simpson, and Fisher indices to evaluate species diversity, and the Chao1 and ACE indices to estimate species richness. Beta diversity, which measures microbiota similarity across groups, was analyzed using weighted and unweighted UniFrac distances. To assess significant differences in beta diversity between groups, a permutational multivariate analysis of variance (PERMANOVA) was performed using the Adonis test. Results: Probiotic supplementation increased Bifidobacterium and Lactobacillus, and decreased Klebsiella and Bacteroides. Beta diversity was significantly altered, while alpha diversity remained unchanged. SCFA levels increased after 7 days. Ammonia levels dropped, and PI values rose. TEER values indicated enhanced barrier integrity. IL-8 and TNF-α decreased, while IL-6 increased. GABA levels remained unchanged. Conclusions: The probiotic combination of Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 modulated gut microbiota composition, metabolic activity, and inflammatory responses in samples from individuals with anxiety on SSRIs, supporting its potential as an adjunctive strategy to mitigate antidepressant-associated dysbiosis. However, limitations—including the small pooled-donor sample, the absence of a healthy control group, and a lack of significant GABA modulation—should be considered when interpreting the findings. Although the SHIME® model is considered a gold standard for microbiota studies, further clinical trials are necessary to confirm these promising results. Full article
Show Figures

Graphical abstract

16 pages, 2613 KiB  
Article
Lingonberry Leaves Modify Rumen Protozoa Population, Carbohydrate Digestion, and Morphology of Gastrointestinal Tract in Sheep: A Preliminary Study
by Małgorzata P. Majewska, Renata Miltko, Grzegorz Bełżecki, Marcin Barszcz, Misza Kinsner and Barbara Kowalik
Molecules 2025, 30(15), 3161; https://doi.org/10.3390/molecules30153161 - 29 Jul 2025
Viewed by 246
Abstract
Leaves, the main by-product of lingonberry harvesting, can be effectively used as a functional feed additive due to their health-promoting properties. This study evaluated the effects of lingonberry leaf (LL) supplementation on rumen fermentation, protozoal populations, and gastrointestinal morphology in sheep. Eight one-year-old [...] Read more.
Leaves, the main by-product of lingonberry harvesting, can be effectively used as a functional feed additive due to their health-promoting properties. This study evaluated the effects of lingonberry leaf (LL) supplementation on rumen fermentation, protozoal populations, and gastrointestinal morphology in sheep. Eight one-year-old Polish Mountain Sheep ewes (mean body weight: 33 kg) were allocated to a control (basal diet; forage-to-concentrate ratio 60:40) or an experimental group (basal diet + 9.30 g/kg DM dried LLs) in a completely randomised design (n = 4 per group) over 34 days. Both diets were formulated to be isoenergetic and isonitrogenous. LL additive significantly reduced Isotrichidae protozoal counts (p < 0.001) and ruminal pectinolytic activity (p = 0.043), without altering short-chain fatty acid (SCFA) or methane concentrations (p > 0.1). Histological analyses showed increased ruminal papilla width and surface area (p < 0.001) and decreased duodenal villus height and muscular layer thickness (p < 0.01). Inflammatory lesions (reddish foci) were identified in the liver in both groups. These findings demonstrate that LL supplementation affected specific protozoal population, fibrolytic activity, and gastrointestinal morphology. Further study on a larger number of animals is recommended to validate the effects and assess the safety and efficacy of LLs as a dietary additive in ruminant nutrition. Full article
(This article belongs to the Special Issue Health Benefits and Applications of Bioactive Phenolic Compounds)
Show Figures

Graphical abstract

30 pages, 10270 KiB  
Article
Fuelling the Fight from the Gut: Short-Chain Fatty Acids and Dexamethasone Synergise to Suppress Gastric Cancer Cells
by Radwa A. Eladwy, Mohamed Fares, Dennis Chang, Muhammad A. Alsherbiny, Chun-Guang Li and Deep Jyoti Bhuyan
Cancers 2025, 17(15), 2486; https://doi.org/10.3390/cancers17152486 - 28 Jul 2025
Viewed by 467
Abstract
Background: Short-chain fatty acids (SCFAs), microbial metabolites also known as postbiotics, are essential for maintaining gut health. However, their antiproliferative effects on gastric cancer cells and potential interactions with conventional therapies remain underexplored. This study aimed to investigate the effects of three SCFA [...] Read more.
Background: Short-chain fatty acids (SCFAs), microbial metabolites also known as postbiotics, are essential for maintaining gut health. However, their antiproliferative effects on gastric cancer cells and potential interactions with conventional therapies remain underexplored. This study aimed to investigate the effects of three SCFA salts—magnesium acetate (A), sodium propionate (P), and sodium butyrate (B)—individually and in combination (APB), as well as in combination with dexamethasone (Dex), on AGS gastric adenocarcinoma cells. Methods: AGS cells were treated with PB, AP, AB, APB, Dex, and APB+Dex. Cell viability was assessed to determine antiproliferative effects, and the IC50 of APB was calculated. Flow cytometry was used to evaluate apoptosis and necrosis. Reactive oxygen species (ROS) levels were measured to assess oxidative stress. Proteomic analysis via LC-MS was performed to identify differential protein expression and related pathways impacted by the treatments. Results: SCFA salts showed significant antiproliferative effects on AGS cells, with APB exhibiting a combined IC50 of 568.33 μg/mL. The APB+Dex combination demonstrated strong synergy (combination index = 0.76) and significantly enhanced growth inhibition. Both APB and APB+Dex induced substantial apoptosis (p < 0.0001) with minimal necrosis. APB alone significantly increased ROS levels (p < 0.0001), while Dex moderated this effect in the combination group APB+Dex (p < 0.0001). Notably, the APB+Dex treatment synergistically targeted multiple tumour-promoting mechanisms, including the impairment of redox homeostasis through SLC7A11 suppression, and inhibition of the haemostasis, platelet activation network and NF-κB signalling pathway via downregulation of NFKB1 (−1.34), exemplified by increased expression of SERPINE1 (1.99) within the “Response to elevated platelet cytosolic Ca2+” pathway. Conclusions: These findings showed a multifaceted anticancer mechanism by APB+Dex that may collectively impair cell proliferation, survival signalling, immune modulation, and tumour microenvironment support in gastric cancer. Full article
(This article belongs to the Special Issue Gut Microbiome, Diet and Cancer Risk)
Show Figures

Figure 1

19 pages, 13401 KiB  
Article
ShenQiGan Extract Repairs Intestinal Barrier in Weaning-Stressed Piglets by Modulating Inflammatory Factors, Immunoglobulins, and Short-Chain Fatty Acids
by Rongxia Guo, Chenghui Jiang, Yanlong Niu, Chun Niu, Baoxia Chen, Ziwen Yuan, Yongli Hua and Yanming Wei
Animals 2025, 15(15), 2218; https://doi.org/10.3390/ani15152218 - 28 Jul 2025
Viewed by 253
Abstract
Weaning stress damages the intestines and disrupts the intestinal barrier in piglets, which significantly impacts the pig farming industry’s economy. We aimed to examine the effects of ShenQiGan extract (CAG) on intestinal barrier function and explore the underlying molecular mechanisms in stress-challenged weaned [...] Read more.
Weaning stress damages the intestines and disrupts the intestinal barrier in piglets, which significantly impacts the pig farming industry’s economy. We aimed to examine the effects of ShenQiGan extract (CAG) on intestinal barrier function and explore the underlying molecular mechanisms in stress-challenged weaned piglets. The experimental design involved 80 weaned piglets aged 28 days (with an average body weight of 7.78 ± 0.074 kg) that were randomly allocated into four groups: Control, LCAG (0.1% CAG), MCAG (0.5% CAG), and HCAG (1.0% CAG). After a 28-day trial period, the growth performance and incidence of diarrhea in piglets were evaluated. CAG increased the average daily gain of weaned piglets, reduced the feed-to-gain ratio, and decreased the incidence of diarrhea. It significantly lowered serum inflammatory cytokine levels while elevating immunoglobulin levels. The supplement notably enhanced concentrations of acetic acid, propionic acid, butyric acid, and isobutyric acid. Furthermore, CAG demonstrated intestinal morphology restoration and upregulation of tight junction proteins and MUC2 protein expression in jejunum. At the mRNA level, it significantly upregulated the expression of Occludin, Claudin1, and MUC2 genes. CAG improves growth performance and mitigates diarrhea in weaned piglets by enhancing intestinal barrier integrity, modulating systemic inflammatory responses, elevating immunoglobulin levels, and promoting short-chain fatty acids (SCFAs) production in the cecum. Full article
(This article belongs to the Section Pigs)
Show Figures

Figure 1

18 pages, 605 KiB  
Review
Gut Microbiota, Microbial Metabolites, and Inflammation in Cardiac Surgery: Implications for Clinical Outcomes—A Narrative Review
by Panagiota Misokalou, Arezina N. Kasti, Konstantinos Katsas and Dimitrios C. Angouras
Microorganisms 2025, 13(8), 1748; https://doi.org/10.3390/microorganisms13081748 - 26 Jul 2025
Viewed by 522
Abstract
Cardiac surgery, particularly procedures involving cardiopulmonary bypass (CPB), is associated with a high risk of postoperative complications, including systemic inflammatory response syndrome (SIRS), postoperative atrial fibrillation (POAF), and infection. Growing evidence suggests that the gut–heart axis, through mechanisms involving intestinal barrier integrity and [...] Read more.
Cardiac surgery, particularly procedures involving cardiopulmonary bypass (CPB), is associated with a high risk of postoperative complications, including systemic inflammatory response syndrome (SIRS), postoperative atrial fibrillation (POAF), and infection. Growing evidence suggests that the gut–heart axis, through mechanisms involving intestinal barrier integrity and gut microbiota homeostasis, may influence these outcomes. This review summarizes the relationship between gut microbiota composition and the inflammatory response in patients undergoing cardiac surgery and the extent to which these alterations impact clinical outcomes. The reviewed studies consistently show that cardiac surgery induces notable alterations in microbial diversity and composition during the perioperative period. These changes, indicative of dysbiosis, are characterized by a reduction in health-associated bacteria such as Blautia, Faecalibacterium, and Bifidobacterium and an increase in opportunistic pathogens. Inflammatory biomarkers were frequently elevated postoperatively, even in patients without evident complications. Key microbial metabolites and biomarkers, including short-chain fatty acids (SCFAs), trimethylamine N-oxide (TMAO), and bile acids (BAs), were implicated in modulating inflammation and clinical outcomes. Additionally, vitamin D deficiency emerged as a contributing factor, correlating with increased systemic inflammation and a higher incidence of POAF. The findings suggest that gut microbiota composition prior to surgery may influence the severity of the postoperative inflammatory response and that perioperative modulation of the gut microbiota could represent a novel approach to improving surgical outcomes. However, the relationship between dysbiosis and acute illness in surgical patients is confounded by factors such as antibiotic use and other perioperative interventions. Large-scale, standardized clinical studies are needed to better define these interactions and guide future therapeutic strategies in cardiac surgery. Full article
Show Figures

Graphical abstract

21 pages, 2411 KiB  
Systematic Review
Response of Akkermansia muciniphila to Bioactive Compounds: Effects on Its Abundance and Activity
by Jair Alejandro Temis-Cortina, Harold Alexis Prada-Ramírez, Hulme Ríos-Guerra, Judith Espinosa-Raya and Raquel Gómez-Pliego
Fermentation 2025, 11(8), 427; https://doi.org/10.3390/fermentation11080427 - 24 Jul 2025
Viewed by 665
Abstract
Introduction: The gut microbiota is vital for human health, and its modulation through dietary and pharmaceutical compounds has gained increasing attention. Among gut microbes, Akkermansia muciniphila has been extensively researched due to its role in maintaining intestinal barrier integrity, regulating energy metabolism, and [...] Read more.
Introduction: The gut microbiota is vital for human health, and its modulation through dietary and pharmaceutical compounds has gained increasing attention. Among gut microbes, Akkermansia muciniphila has been extensively researched due to its role in maintaining intestinal barrier integrity, regulating energy metabolism, and influencing inflammatory responses. Subject: To analyze and synthesize the available scientific evidence on the influence of various bioactive compounds, including prebiotics, polyphenols, antioxidants, and pharmaceutical agents, on the abundance and activity of A. muciniphila, considering underlying mechanisms, microbial context, and its therapeutic potential for improving metabolic and intestinal health. Methods: A systematic literature review was conducted in accordance with the PRISMA 2020 guidelines. Databases such as PubMed, ScienceDirect, Scopus, Web of Science, SciFinder-n, and Google Scholar were searched for publications from 2004 to 2025. Experimental studies in animal models or humans that evaluated the impact of bioactive compounds on the abundance or activity of A. muciniphila were prioritized. The selection process was managed using the Covidence platform. Results: A total of 78 studies were included in the qualitative synthesis. This review compiles and analyzes experimental evidence on the interaction between A. muciniphila and various bioactive compounds, including prebiotics, antioxidants, flavonoids, and selected pharmaceutical agents. Factors such as the chemical structure of the compounds, microbial environment, underlying mechanisms, production of short-chain fatty acids (SCFAs), and mucin interactions were considered. Compounds such as resistant starch type 2, GOS, 2′-fucosyllactose, quercetin, resveratrol, metformin, and dapagliflozin showed beneficial effects on A. muciniphila through direct or indirect pathways. Discussion: Variability across studies reflects the influence of multiple variables, including compound type, dose, intervention duration, experimental models, and analytical methods. These differences emphasize the need for a contextualized approach when designing microbiota-based interventions. Conclusions: A. muciniphila emerges as a promising therapeutic target for managing metabolic and inflammatory diseases. Further mechanistic and clinical studies are necessary to validate its role and to support the development of personalized microbiota-based treatment interventions. Full article
(This article belongs to the Section Probiotic Strains and Fermentation)
Show Figures

Figure 1

Back to TopTop