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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (835)

Search Parameters:
Keywords = fecal fatty acids

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 8603 KB  
Article
Metagenomics Investigation on Baby Diaper Area Microbiome and Its Association with Skin pH and Dermatitis in the Diapered Area
by Ping Hu, Andrew N. Carr, Mirjana Parlov, Dionne Swift, Jay P. Tiesman, Nivedita Ramji, Jennifer J. Schoch and Amber G. Teufel
Microorganisms 2025, 13(11), 2632; https://doi.org/10.3390/microorganisms13112632 - 20 Nov 2025
Abstract
Dermatitis in the diapered area (DDA) is the most common skin condition in infants and can cause significant pain and discomfort, leading to disturbed sleep, changes in temperament, and heightened concern and anxiety for caregivers. This study investigates the relationship between skin pH, [...] Read more.
Dermatitis in the diapered area (DDA) is the most common skin condition in infants and can cause significant pain and discomfort, leading to disturbed sleep, changes in temperament, and heightened concern and anxiety for caregivers. This study investigates the relationship between skin pH, microbiome composition, and DDA severity in 158 infants from China, the US, and Germany, focusing on the buttocks, perianal, and thigh regions. Significant variations in DNA biomass and microbiota profiles were noted. Escherichia coli and Veillonella atypica were linked to higher rash scores and elevated skin pH, while Bifidobacterium longum showed a negative correlation with buttocks pH and rash severity but not with perianal rash. Correlation patterns emerged for other species, like Enterococcus faecalis, between perianal and buttocks rashes. Functional analysis identified key categories, including lipid and fatty acid metabolism, cofactor, amino acid, and carbohydrate metabolism, homeostasis and osmolarity stress, and microbial virulence and oxidative stress response, which are vital for skin health, DDA, and pH regulation in infants. These findings underscore the importance of maintaining a mildly acidic skin pH and minimizing fecal and urine residues for optimal infant skin health, suggesting that microbiota significantly influence DDA development, and provide insights for future preventive strategies and therapeutic interventions. Full article
(This article belongs to the Special Issue Skin Microbiome)
Show Figures

Figure 1

17 pages, 7667 KB  
Article
Peimine Alleviates DSS-Induced Colitis by Modulating Gut Microbiota and Attenuating Inflammation and Oxidative Stress
by Xuke Guan, Deping Han, Haojie Sha, Moyue Yao, Jiaying Zhang, Guangyao Zhang, Yibing Wu, Dingding Su and Qing Yang
Int. J. Mol. Sci. 2025, 26(22), 11203; https://doi.org/10.3390/ijms262211203 - 20 Nov 2025
Abstract
Peimine (PM), a steroidal alkaloid derived from aged garlic (Allium sativum L.), demonstrates potent therapeutic efficacy against ulcerative colitis (UC) through multi-target mechanisms. Integrating network pharmacology and in vivo validation, we reveal that PM suppresses colitis by concurrently inhibiting PI3K-AKT, JAK-STAT, and [...] Read more.
Peimine (PM), a steroidal alkaloid derived from aged garlic (Allium sativum L.), demonstrates potent therapeutic efficacy against ulcerative colitis (UC) through multi-target mechanisms. Integrating network pharmacology and in vivo validation, we reveal that PM suppresses colitis by concurrently inhibiting PI3K-AKT, JAK-STAT, and HIF-1 signaling pathways—key drivers of inflammation and oxidative stress. In a murine model of dextran sulfate sodium (DSS)-induced UC, oral PM administration (4 mg/kg) significantly attenuated disease severity, evidenced by reduced disease activity index, restored colon length, and improved epithelial barrier integrity. PM treatment diminished pro-inflammatory cytokines TNF-α (4.2-fold) and IL-6 (3.1-fold) and oxidative damage while reshaping gut microbiota composition to enrich beneficial taxa (Akkermansia muciniphila, Lactobacillus spp.). Critically, PM rescued fecal short-chain fatty acid (SCFA) production (acetate, propionate, butyrate), directly linking microbial remodeling to mucosal healing. These findings establish PM as a novel natural compound targeting inflammation-redox-microbiota crosstalk, offering a promising pharmacological strategy for UC management. Full article
(This article belongs to the Section Molecular Microbiology)
Show Figures

Figure 1

16 pages, 1267 KB  
Review
The Role of Microbiota in Ovarian Cancer: Implications for Treatment Response and Therapeutic Strategies
by Jose-Ramon Blanco, Rosa del Campo, José Avendaño-Ortiz, Mariano Laguna-Olmos and Amancio Carnero
Cells 2025, 14(22), 1813; https://doi.org/10.3390/cells14221813 - 19 Nov 2025
Viewed by 31
Abstract
Cancer remains a global health challenge (18.1 million new cases in 2020), with incidence projected to reach 28 million within two decades. Ovarian cancer (OC) is the deadliest gynecologic malignancy, usually diagnosed at advanced stages and with poorly understood etiology. Emerging evidence implicates [...] Read more.
Cancer remains a global health challenge (18.1 million new cases in 2020), with incidence projected to reach 28 million within two decades. Ovarian cancer (OC) is the deadliest gynecologic malignancy, usually diagnosed at advanced stages and with poorly understood etiology. Emerging evidence implicates reproductive tract and gut microbiota in OC biology. Microbiota shape carcinogenesis via turnover, immunity, and metabolism; dysbiosis promotes DNA damage, inflammation, and carcinogenic metabolites, engaging multiple hallmarks of cancer. In OC, microbes may reach tumors by local ascent, translocation, or hematogenous spread, originating from vagina, upper reproductive tract, peritoneal fluid, or gut. Lactobacillus-dominant vaginal communities support mucosal integrity, whereas anaerobes disrupt barriers, increase inflammation, and correlate with OC risk; mouse models show vaginal dysbiosis accelerates tumor progression. Distinct microbial profiles in upper reproductive sites and peritoneal fluid associated with immune remodeling. Gut dysbiosis drives barrier loss, immune imbalance, and estrogen reactivation. Microbial metabolites (lipopolysaccharides, short-chain fatty acids) modulate oncogenic pathways, altering epithelial–mesenchymal transition, immune evasion, and drug resistance. Across cohorts, OC tissues and fluids show Pseudomonadota/Bacteroidota enrichment and Akkermansia depletion; fecal microbiota from OC patients accelerates tumor growth in mice, whereas Akkermansia supplementation restores antitumor immunity. Antibiotic exposure and platinum resistance associate with reduced diversity and expansion of lactate-producing taxa. Microbiome-informed interventions–diet, probiotics/postbiotics, fecal microbiota transfer, and selective antibiotics–may augment chemotherapy and immunotherapy. Overall, the microbiome is a modifiable determinant of OC risk, progression, and treatment response, warranting rigorous, standardized, multi-omics studies. Full article
Show Figures

Figure 1

27 pages, 1272 KB  
Review
The Pediatric Microbiota–Gut–Brain Axis: Implications for Neuropsychiatric Development and Intervention
by Giuseppe Marano, Greta Sfratta, Ester Maria Marzo, Giorgia Cozzo, Francesca Abate, Gianandrea Traversi, Osvaldo Mazza, Esmeralda Capristo, Eleonora Gaetani and Marianna Mazza
Children 2025, 12(11), 1561; https://doi.org/10.3390/children12111561 - 17 Nov 2025
Viewed by 353
Abstract
Background: The gut microbiota plays a crucial role in brain development and function, especially in early life. Disruptions in the pediatric microbiota–gut–brain axis have been linked to neurodevelopmental and psychiatric disorders. We hypothesize that early-life dysbiosis can perturb neurodevelopment via the pediatric microbiota–gut–brain [...] Read more.
Background: The gut microbiota plays a crucial role in brain development and function, especially in early life. Disruptions in the pediatric microbiota–gut–brain axis have been linked to neurodevelopmental and psychiatric disorders. We hypothesize that early-life dysbiosis can perturb neurodevelopment via the pediatric microbiota–gut–brain axis, increasing risk and/or severity of neuropsychiatric outcomes, and that microbiota-targeted strategies may mitigate this risk. Methods: We conducted a narrative review by searching PubMed, Scopus, and Web of Science up to January 2025 for studies addressing pediatric microbiota, neuropsychiatric development, and interventions. Human and animal studies were included if they provided mechanistic or clinical insights. Results: Key determinants of microbiota development in childhood include mode of delivery, feeding practices, antibiotic exposure, diet, and environment. Altered microbial composition has been associated with autism spectrum disorder, attention-deficit/hyperactivity disorder, mood disorders, anxiety, and anorexia nervosa. Mechanistic pathways involve immune modulation, neural signaling (including the vagus nerve and enteric nervous system), and microbial metabolites such as short-chain fatty acids. Interventions targeting the microbiota—ranging from dietary strategies and probiotics to psychobiotics and fecal microbiota transplantation—show promise but require further pediatric-focused trials. Conclusions: The pediatric microbiota–gut–brain axis represents a critical window for neuropsychiatric vulnerability and intervention. Early-life strategies to support a healthy microbiota may help reduce the risk or severity of psychiatric disorders. Future research should prioritize longitudinal pediatric cohorts and clinical trials to translate mechanistic insights into precision interventions. Full article
Show Figures

Figure 1

20 pages, 1442 KB  
Systematic Review
Short-Chain Fatty Acids and Colorectal Cancer: A Systematic Review and Integrative Bayesian Meta-Analysis of Microbiome–Metabolome Interactions and Intervention Efficacy
by Yingge He, Ke Peng, Junze Tan, Yonghui Hao, Shiyan Zhang, Changqing Gao and Liqi Li
Nutrients 2025, 17(22), 3552; https://doi.org/10.3390/nu17223552 - 14 Nov 2025
Viewed by 455
Abstract
Objective: Existing studies on short-chain fatty acids (SCFAs) and colorectal cancer (CRC) yield contradictory conclusions and are limited to single ethnic groups or sample types. This study aimed to (1) quantify associations between total SCFAs/subtypes (acetate, propionate, butyrate) and CRC/advanced colorectal adenoma [...] Read more.
Objective: Existing studies on short-chain fatty acids (SCFAs) and colorectal cancer (CRC) yield contradictory conclusions and are limited to single ethnic groups or sample types. This study aimed to (1) quantify associations between total SCFAs/subtypes (acetate, propionate, butyrate) and CRC/advanced colorectal adenoma (A-CRA) risks; (2) identify modifiers (ethnicity, sample type, intervention); and (3) clarify SCFA–gut microbiota interaction mechanisms via integrative Bayesian meta-analysis and multi-ancestry data integration. Methods: We systematically searched PubMed, Embase, Cochrane Library, and Web of Science (inception to September 2025) using keywords: “Short-chain fatty acids”, “SCFAs”, “Colorectal cancer”, “CRC”, “Gut microbiota”, “Dietary fiber”, and “High-amylose maize starch butyrate”. Eligible studies included 14 peer-reviewed original studies (7 observational, cohort/case–control/cross-sectional; 7 RCTs) covering Europeans, Asians, and African Americans. Inclusion criteria: Quantitative SCFA data (total/≥3 subtypes), clear ethnic grouping, reported CRC/A-CRA risks or intervention outcomes. Exclusion criteria: Reviews, animal/in vitro studies, incomplete data, low-quality studies (Newcastle–Ottawa Scale [NOS] <6 for observational; high Cochrane risk for RCTs), or limited populations (single gender/rare genetics). A Bayesian hierarchical random-effects model quantified effect sizes (Odds Ratio [OR]/Mean Difference [MD], 95% credible intervals [CrI]), with heterogeneity analyzed via multi-ancestry stratification, intervention efficacy, and microbiota interaction analyses (Preferred Reporting Items for Systematic Reviews and Meta-Analyses [PRISMA] 2020; International Prospective Register of Systematic Reviews [PROSPERO]: CRD420251157250). Results: Total SCFAs were negatively associated with CRC (OR = 0.78, 95% CrI: 0.65–0.92) and A-CRA (OR = 0.72, 95% CrI: 0.59–0.87), with butyrate showing the strongest protective effect (CRC: OR = 0.63, 95% CrI: 0.51–0.77). Ethnic heterogeneity was significant: Europeans had the strongest protection (OR = 0.71), Asians had weaker protection (OR = 0.86), and African Americans had the lowest fecal SCFA levels and the highest CRC risk. Fecal SCFAs showed a stronger CRC association than serum/plasma SCFAs (OR = 0.73 vs. 0.85). High-Amylose Maize Starch Butyrate (HAMSB) outperformed traditional fiber in increasing fecal butyrate (MD = 4.2 mmol/L vs. 2.8 mmol/L), and high butyrate-producing bacteria (Clostridium, Roseburia) enhanced SCFA protection (OR = 0.52 in high-abundance groups). Conclusions: SCFAs (especially butyrate) protect against CRC and precancerous lesions, with effects modulated by ethnicity, sample type, and gut microbiota. High-Amylose Maize Starch Butyrate is a priority intervention for high-risk populations (e.g., familial adenomatous polyposis, FAP), and differentiated strategies are needed: 25–30 g/d dietary fiber for Europeans, 20–25 g/d for Asians, and probiotics (Clostridium) for African Americans. Future Perspectives: Expand data on underrepresented groups (African Americans, Latinos), unify SCFA detection methods, and conduct long-term RCTs to validate intervention efficacy and “genetics-microbiota-metabolism” crosstalk—critical for CRC precision prevention. Full article
(This article belongs to the Section Lipids)
Show Figures

Figure 1

21 pages, 1813 KB  
Review
The Microbiota–Gut–Brain Axis in Autism: Associations, Causal Inference, and Interventions—A Narrative Review
by Zhiqiang Zhang, Wenkai Kang, Yu Mi, Xin Zhong and Yulong He
Pathogens 2025, 14(11), 1145; https://doi.org/10.3390/pathogens14111145 - 11 Nov 2025
Viewed by 551
Abstract
Autism spectrum disorder is markedly heterogeneous and frequently accompanied by gastrointestinal symptoms that often correlate with behavioral phenotypes. Emerging evidence suggests that the microbiota–gut–brain axis may contribute to these associations through multiple bidirectional communication routes—including neural, immune, and endocrine pathways, as well as [...] Read more.
Autism spectrum disorder is markedly heterogeneous and frequently accompanied by gastrointestinal symptoms that often correlate with behavioral phenotypes. Emerging evidence suggests that the microbiota–gut–brain axis may contribute to these associations through multiple bidirectional communication routes—including neural, immune, and endocrine pathways, as well as microbial metabolites such as short-chain fatty acids and tryptophan–kynurenine intermediates. This narrative review synthesizes clinical, mechanistic, and interventional evidence published between January 2010 and July 2025, clarifies the extent to which current data support association versus causation, evaluates key confounding factors, summarizes evidence for interventions such as probiotics, prebiotics, and fecal microbiota transplantation, and outlines future directions for precision research and targeted interventions based on functional pathways and stratified subgroups. Full article
Show Figures

Figure 1

19 pages, 8725 KB  
Article
Postbiotics from Lacticaseibacillus rhamnosus IOB820 Combat Obesity in HFD Mice by Modulating Gut Microbiota and Enhancing SCFA Production
by Xiaomin Feng, Hanlu Li, Jianxia Tian, Xuemei Han, Wu Liang, Feiliang Zhong and Xuegang Luo
Nutrients 2025, 17(22), 3525; https://doi.org/10.3390/nu17223525 - 11 Nov 2025
Viewed by 285
Abstract
Aims: To evaluate whether Lacticaseibacillus rhamnosus (L. rhamnosus) IOB820 and its postbiotics can combat high-fat diet (HFD)-induced obesity, improve metabolic parameters, and modulate gut microbiota and systemic inflammation in a mouse model. Methods: Seventy 4-week-old male C57BL/6J mice were divided into [...] Read more.
Aims: To evaluate whether Lacticaseibacillus rhamnosus (L. rhamnosus) IOB820 and its postbiotics can combat high-fat diet (HFD)-induced obesity, improve metabolic parameters, and modulate gut microbiota and systemic inflammation in a mouse model. Methods: Seventy 4-week-old male C57BL/6J mice were divided into a normal diet group, an HFD control group, two postbiotic dose groups, two live bacteria dose groups, and an orlistat control group. After 10 weeks of intervention with live L. rhamnosus IOB820 or its postbiotics, body weight, metabolic parameters (blood glucose, lipid profile, hepatic steatosis), pro- and anti-inflammatory cytokines (TNF-α, IL-6, IL-1β, IL-10), gut microbiota composition (α, β diversity and taxonomic shifts), and fecal short-chain fatty acid (SCFA) levels were assessed. Results: Both live L. rhamnosus IOB820 and its postbiotics significantly alleviated HFD-induced weight gain and improved metabolic outcomes. The treatments also reduced systemic inflammation, as indicated by decreased levels of TNF-α, IL-6, and IL-1β and elevated IL-10. These effects were accompanied by restoration of gut microbial diversity, enrichment of beneficial taxa, and increased fecal SCFA concentrations. Conclusions: L. rhamnosus IOB820 and its postbiotics effectively mitigate obesity and related metabolic disturbances in HFD-fed mice. Their beneficial effects are likely mediated through modulation of gut microbiota composition and enhancement of SCFA-driven anti-inflammatory responses. Full article
(This article belongs to the Special Issue Fermented Foods and Health Modulation)
Show Figures

Figure 1

29 pages, 16647 KB  
Article
A Specific Ratio of Dietary Short-Chain and Long-Chain Fructo-Oligosaccharides Shifts the Immune Response Away from Type 2 in a Murine Model for House Dust Mite-Induced Asthma
by Roos E. M. Verstegen, Marit Zuurveld, Suzan Thijssen, Marjolein J. W. de Bruijn, Ingrid van Ark, Mara A. P. Diks, Johan Garssen, Gert Folkerts, Atanaska I. Kostadinova, Rudi W. Hendriks and Linette E. M. Willemsen
Nutrients 2025, 17(22), 3520; https://doi.org/10.3390/nu17223520 - 11 Nov 2025
Viewed by 275
Abstract
Background/Objectives: The gut microbiome has an important role in immune regulation, and dietary interventions that support a balanced microbiota may help to prevent the development of allergic asthma. Dietary fibers can beneficially affect the intestinal microbiome, but due to the diversity of [...] Read more.
Background/Objectives: The gut microbiome has an important role in immune regulation, and dietary interventions that support a balanced microbiota may help to prevent the development of allergic asthma. Dietary fibers can beneficially affect the intestinal microbiome, but due to the diversity of fiber types, the effects differ. In this study, we investigate the preventive effects of two mixes of short-chain and long-chain (1:1 and 9:1 ratio) fructo-oligosaccharides (FOS) in a mouse model of house dust mite (HDM)-induced allergic asthma. Methods: BALB/c mice received FOS-supplemented (1% w/w) diets before and during intranasal exposures to HDM. Endpoint airway hyperreactivity measurements were performed, followed by the collection of bronchoalveolar lavage fluid (BALF), lung, serum and cecum content. Fecal microbiome composition was determined by DNA sequencing and short-chain fatty acid (SCFA) levels were determined in the cecum, serum and lung. Results: Fecal microbiome analyses revealed an increased abundance of Prevotellaceae after FOS1:1 supplementation in HDM-allergic mice. Additionally, FOS1:1 protected against an HDM-induced increase in basal airway resistance. Both FOS1:1 and FOS9:1 restored the systemic acetate levels in HDM-allergic mice. The two FOS supplementations did not affect HDM-induced inflammatory cell influx in the BALF. However, FOS1:1 increased the frequency of Th1-cells and prevented an HDM-induced increase in the Th2/Th1 balance. Upon ex vivo restimulation with HDM, lung cell suspensions of FOS1:1-fed mice produced less type 2-related cytokines compared to control-supplemented mice, and FOS9:1 followed a similar pattern. Conclusions: Specific short-chain and long-chain FOS ratios differentially affect the microbiome and immune system in a mouse model with HDM-induced allergic airway inflammation. Dietary supplementation with FOS1:1 shifts the immune response away from type 2, suggesting that dietary fibers like FOS1:1 may contribute as a part of a broader strategy to modulate HDM-induced allergic asthma. Full article
Show Figures

Figure 1

20 pages, 5932 KB  
Article
Xanthoceras sorbifolium Bunge Leaves Ameliorate Type 2 Diabetes Mellitus by Modulating Glucolipid Metabolism Through the Gut Microbiota–Metabolite Axis
by Qiong Jia, Xianyu Zhang, Mengting Han, Tian Zhong and Hui Zhou
Foods 2025, 14(22), 3809; https://doi.org/10.3390/foods14223809 - 7 Nov 2025
Viewed by 462
Abstract
Xanthoceras sorbifolium Bunge leaves (XBL), traditionally consumed as herbal tea, have attracted increasing attention as potential functional food ingredients for managing type 2 diabetes mellitus (T2DM). This study investigated the anti-diabetic effects of an aqueous XBL extract in T2DM rats induced with a [...] Read more.
Xanthoceras sorbifolium Bunge leaves (XBL), traditionally consumed as herbal tea, have attracted increasing attention as potential functional food ingredients for managing type 2 diabetes mellitus (T2DM). This study investigated the anti-diabetic effects of an aqueous XBL extract in T2DM rats induced with a high-fat, high-sucrose diet combined with streptozotocin. XBL administration significantly improved glycemic control, insulin sensitivity, lipid profiles, and pancreatic and renal histopathology. Integrated 16S rRNA sequencing and untargeted fecal metabolomics revealed the modulation of key metabolic pathways, including linoleic acid and histidine metabolism, and elevated production of short-chain fatty acids (SCFAs) such as acetate and propionate. XBL also enriched beneficial gut microbes including Prevotella, Lachnospiraceae_NK4A136_group, and [Eubacterium]_xylanophilum_group, whose abundance showed positive correlations with SCFA levels and metabolic improvements. These findings demonstrate that XBL ameliorates T2DM through gut microbiota–SCFA–metabolite interactions and suggest its potential as a natural, multi-target dietary strategy for metabolic health management. Full article
Show Figures

Graphical abstract

19 pages, 878 KB  
Article
In Vitro Influence of a Chemically Characterized Hippophae rhamnoides L. Fruit Extract on Healthy and Constipated Human Gut Microbiota Functionality and Aquaporin-3 Expression
by Lorenza Francesca De Lellis, Ángela Toledano-Marín, Miguel Navarro-Moreno, Elisabetta Caiazzo, Gennaro Madonna, Adriana Delgado-Osorio, Daniele Giuseppe Buccato, Luana Izzo, Antonio Paolillo, Alessandro Di Minno, Hammad Ullah, Maria Vittoria Morone, Anna De Filippis, Massimiliano Galdiero, Armando Ialenti, José Ángel Rufián Henares and Maria Daglia
Foods 2025, 14(21), 3800; https://doi.org/10.3390/foods14213800 - 6 Nov 2025
Viewed by 484
Abstract
To identify the underlying mechanisms by which H. rhamnoides fruit extract exerts regulatory effects on intestinal function, we investigated its chemical composition using UHPLC Q-Orbitrap HRMS and evaluated its biological effects on Aquaporin-3 (AQP-3) expression via Western blot in the intestinal epithelial cell [...] Read more.
To identify the underlying mechanisms by which H. rhamnoides fruit extract exerts regulatory effects on intestinal function, we investigated its chemical composition using UHPLC Q-Orbitrap HRMS and evaluated its biological effects on Aquaporin-3 (AQP-3) expression via Western blot in the intestinal epithelial cell line (HT-29). Moreover, fecal microbiota from healthy and constipated adults was employed to mimic the in vitro fermentation of the digested extract and evaluate its effects on gut microbiota functionality. Antioxidant capacity (i.e., Total Phenolic Contents (TPC), 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS), 2,2-diphenyl-1-picrylhydrazyl (DPPH), and ferric reducing antioxidant power (FRAP) assays) was assessed prior to and after simulated digestion and fermentation processes. Short-chain fatty acids (SCFAs) were quantified using UHPLC-RID of the fermented samples. In the extract, 23 compounds belonging to a variety of classes (mainly polyphenols) were tentatively identified. The extract significantly upregulated AQP-3 expression in the absence of cytotoxicity. After in vitro fermentation with gut microbiota isolated from constipated subjects, ABTS and FRAP values significantly decreased, as well as TPC, suggesting a greater consumption of antioxidant compounds, consistent with the increased production of radical compounds associated with constipation. Fermentation with intestinal microbiota with healthy and constipated gut microbiota resulted in an increase in SCFA. These results provide preliminary insights into a non-pharmacological strategy for functional constipation. Full article
(This article belongs to the Section Plant Foods)
Show Figures

Figure 1

18 pages, 4262 KB  
Article
Multi-Omics Insights into the Impact of Fermented Wheat Bran-Soybean Meal-Broussonetia papyrifera Mixture Substance on the Gut Microbiota of Late Gestation Sows In Vitro
by Lele Fu, Yushi Chen, Yantao Li and Cheng Wang
Animals 2025, 15(21), 3199; https://doi.org/10.3390/ani15213199 - 3 Nov 2025
Viewed by 264
Abstract
Comprehensive maternal nutritional interventions, particularly during late gestation, enhance perinatal outcomes and support long-term maternal-offspring health by modulating the microbiota. Fermented diets are recommended for inclusion in dietary guidelines during gestation, yet the specific metabolites after fermentation and their specific regulatory effects on [...] Read more.
Comprehensive maternal nutritional interventions, particularly during late gestation, enhance perinatal outcomes and support long-term maternal-offspring health by modulating the microbiota. Fermented diets are recommended for inclusion in dietary guidelines during gestation, yet the specific metabolites after fermentation and their specific regulatory effects on gut microbiota during late gestation remain unclear. This study investigates the functional benefits of a fermented wheat bran–soybean meal–Broussonetia papyrifera mixed substrate (FMS) on the late-gestation gut microbiota using an in vitro fermentation model. The FMS was first fermented for 72 h with bacterial and enzymatic agents (2% v/v), then anaerobically incubated with fecal inocula from Jinhua pigs. Fermentation significantly enhanced nutritional profiles, increasing crude protein and amino acids while reducing fiber components (neutral detergent fiber, acid detergent fiber, and non-starch polysaccharide, p < 0.05). Metabolome analysis revealed a significant increase in the abundance of organic acids, amino acids, and short peptides in FMS, along with the enrichment of D-amino acid and sphingolipid pathways (p < 0.05). In addition, FMS significantly increased the abundance of Limosilactobacillus and Lactobacillus, as well as short-chain fatty acids production, compared to the unfermented group (p < 0.05). These findings demonstrate that fermentation pretreatment reduces fiber components, enhances flavor compounds and bioactive metabolites, thereby optimizing microbial utilization and increasing short-chain fatty acids production. Full article
(This article belongs to the Collection The Weaned Pig: Nutrition and Management)
Show Figures

Figure 1

16 pages, 3063 KB  
Article
Alterations in Serum Immune Parameters, Cytokines, Intestinal Permeability, Fecal Microbiota, and Short-Chain Fatty Acids in Healthy and Diarrheic Suckling Calves
by Peiyun Gao, Shaoyang Pang, Yaqin Tang, Qianqian Wang, Qiuyan Li, Wenju Zhang, Cunxi Nie, Junli Niu and Kexun Lian
Agriculture 2025, 15(21), 2289; https://doi.org/10.3390/agriculture15212289 - 3 Nov 2025
Viewed by 390
Abstract
This study compared serum immunological parameters, cytokines, intestinal permeability, fecal microbiota, and short-chain fatty acids (SCFAs) between healthy and diarrheic suckling calves. Serum and facecal samples were analyzed using ELISA kits, 16S rDNA sequencing, and targeted metabolomics. Compared with healthy calves, the serum [...] Read more.
This study compared serum immunological parameters, cytokines, intestinal permeability, fecal microbiota, and short-chain fatty acids (SCFAs) between healthy and diarrheic suckling calves. Serum and facecal samples were analyzed using ELISA kits, 16S rDNA sequencing, and targeted metabolomics. Compared with healthy calves, the serum levels of aspartate aminotransferase, interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), endotoxin (ET), and diamine oxidase (DAO) were significantly higher (p < 0.05), whereas the serum levels of immunoglobulin A (IgA), immunoglobulin G (IgG), and interleukin-10 (IL-10) were significantly lower in diarrheic calves (p < 0.05). The contents of propionic acid, butyric acid, and valeric acid significantly decreased in the fecal of diarrheic calves (p < 0.05). Moreover, the Chao1 and observed_features index of fecal microbiota significantly decreased in diarrheic calves (p < 0.05). The relative abundance of Escherichia-Shigella, Clostridium_sensu_stricto_1, and Streptococcus was significantly higher (p < 0.05), whereas Phascolarctobacterium, Ruminococcus torques group, and Faecalibacterium were significantly lower in diarrheic calves (p < 0.05). Escherichia-Shigella abundance was positively correlated with ET, DAO, IL-1β, and TNF-α levels (p < 0.05). Faecalibacterium abundance was significantly positively correlated with IgG, IgA, IL-10, and butyric acid but negatively correlated with ET and DAO levels (p < 0.05). In summary, diarrheic suckling calves exhibited reduced immune function, inflammatory response, and increased intestinal permeability. The relative abundance of fecal microbiota of Escherichia-Shigella and Clostridium_sensu_stricto_1 increased, while propionic acid, butyric acid, and valeric acid concentration were decreased in calves with diarrhea. This underscores the critical interplay between microbiota balance and gut health in diarrhea. Full article
(This article belongs to the Special Issue Research on the Nutrition and Physiology of Dairy and Beef Cattle)
Show Figures

Figure 1

16 pages, 3576 KB  
Article
Computational Analysis of the Effect of Dietary Interventions on the Gut Microbiome Composition in Parkinson’s Disease
by López Franyer, García Macias Adrian, Beltran Oscar, González Janneth and Pinzón Andrés
Bacteria 2025, 4(4), 59; https://doi.org/10.3390/bacteria4040059 - 3 Nov 2025
Viewed by 350
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder characterized by motor symptoms like tremor, rigidity, and bradykinesia. The WHO estimates that 10 million people currently have PD, with its prevalence expected to double to 20 million by 2050. Key risk factors include age, male [...] Read more.
Parkinson’s disease (PD) is a neurodegenerative disorder characterized by motor symptoms like tremor, rigidity, and bradykinesia. The WHO estimates that 10 million people currently have PD, with its prevalence expected to double to 20 million by 2050. Key risk factors include age, male sex, environmental contaminants, and family history. Emerging evidence links gut microbiota dysbiosis to PD, suggesting it contributes to neuroinflammation and disease progression, though the role of dietary interventions remains unclear. This study used computational simulations with genome-scale metabolic models (GEMs) to analyze how diet impacts the gut microbiota in PD patients. Fecal microbiota from PD patients and healthy controls were compared across three diets: high-fiber, Mediterranean, and vegan. Simulations revealed increased pro-inflammatory bacteria (e.g., Escherichia coli O157) in PD patients, likely due to reduced bacterial competition, alongside the decreased production of beneficial metabolites like butyrate, phenylalanine, and cysteine. The Mediterranean diet showed higher short-chain fatty acid production, potentially benefiting PD patients. These findings underscore the importance of dietary interventions in modulating the gut microbiome and suggest that targeted diets may complement PD therapies, improving patient outcomes. Full article
Show Figures

Figure 1

18 pages, 1203 KB  
Article
Exploratory In Vitro Evaluation of Maternal–Infant Bifidobacterium Strains for Microbiota Modulation in a Pediatric Cystic Fibrosis Context
by Maria Esteban-Torres, Isabel Blanco, Andrea Asensio-Grau, Nuria Ruiz, Manuel Bernabeu and Joaquim Calvo-Lerma
Microorganisms 2025, 13(11), 2523; https://doi.org/10.3390/microorganisms13112523 - 2 Nov 2025
Viewed by 500
Abstract
This study explores the potential of novel Bifidobacterium isolates as targeted probiotic supplements for children with cystic fibrosis (CF), a condition often associated with gut dysbiosis. Five strains of Bifidobacterium genus (B. animalis IATA01, B. pseudocatenulatum IATA35, B. longum [...] Read more.
This study explores the potential of novel Bifidobacterium isolates as targeted probiotic supplements for children with cystic fibrosis (CF), a condition often associated with gut dysbiosis. Five strains of Bifidobacterium genus (B. animalis IATA01, B. pseudocatenulatum IATA35, B. longum IATA02, B. bifidum IATA13, and B. longum IATA05) isolated from maternal–infant fecal samples were assessed in vitro following the FAO/WHO guidelines. Their probiotic potential was evaluated through simulated gastrointestinal digestion in the CF context, their adhesion to mucin, and their carbohydrate fermentation capacity. Additionally, their impact on colonic microbiota modulation was analyzed using static in vitro colonic fermentation with fecal inocula from four pediatric patients with CF to assess the presence of different bacterial groups associated with dysbiosis via qPCR and short-chain fatty acid production by GC-MS. Three strains (IATA01, IATA35, and IATA05) demonstrated survival after gastrointestinal digestion, with IATA01 exhibiting the highest adhesion to mucin but limited carbohydrate fermentation capacity. All strains increased the Bifidobacterium levels after colonic fermentation, while their effects on reducing pathogenic groups and promoting beneficial bacteria such as Akkermansia and Faecalibacterium varied depending on the strain and the individual inoculum. These findings highlight the strain-specific effects of Bifidobacterium and evidence a specific impact on colonic microbiota, depending on the composition of the basal inoculum, highlighting individual-specific responses. Full article
(This article belongs to the Section Gut Microbiota)
Show Figures

Figure 1

37 pages, 1558 KB  
Review
Gut Microbiota: An Ally in the Mechanisms and Interventions of Healthy Aging
by Samia Chatterjee, Ananda Vardhan Hebbani and Khajamohiddin Syed
Gastrointest. Disord. 2025, 7(4), 68; https://doi.org/10.3390/gidisord7040068 - 26 Oct 2025
Viewed by 1176
Abstract
The gut microbiota greatly influences host physiology, including immune regulation, metabolic balance, and brain health. Aging is associated with alterations in the gut microbiome, including reduced microbial diversity and increased pro-inflammatory bacteria, which are linked to age-related decline and chronic diseases. This review [...] Read more.
The gut microbiota greatly influences host physiology, including immune regulation, metabolic balance, and brain health. Aging is associated with alterations in the gut microbiome, including reduced microbial diversity and increased pro-inflammatory bacteria, which are linked to age-related decline and chronic diseases. This review examines the impact of the gut microbiota on key indicators of aging, including cellular senescence, mitochondrial dysfunction, alterations in gene expression, and immune system modifications. It also examines microbiome-related diseases associated with aging, including neurodegeneration, cardiovascular issues, metabolic syndrome, and frailty. Additionally, it highlights evidence-based methods to restore a youthful microbial profile. New findings suggest that certain microbial substances, including short-chain fatty acids, urolithins, and bile acids, play a role in regulating inflammation, maintaining barrier integrity, and influencing metabolism. Age-related diseases are often associated with molecular pathways driven by an imbalance in the gut microbiome. Various intervention strategies, from dietary changes and probiotics to personalized nutrition and fecal microbiota transplantation, have shown promise in reversing signs of microbial aging and improving health outcomes in both lab and human studies. Overall, the gut microbiome serves as both a marker and a regulator of healthy aging. Treatments that restore microbial balance offer hopeful ways to extend healthy living. Future studies should focus on developing long-term, multifaceted, and personalized methods to identify causal pathways and enhance microbiota-based strategies for various aging populations. Full article
Show Figures

Figure 1

Back to TopTop