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

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

Search Results (907)

Search Parameters:
Keywords = mucosal inflammation

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 4046 KiB  
Article
TMAO Activates the NLRP3 Inflammasome, Disrupts Gut–Kidney Interaction, and Promotes Intestinal Inflammation
by Leyao Fang, Junxi Shen, Nenqun Xiao and Zhoujin Tan
Int. J. Mol. Sci. 2025, 26(15), 7441; https://doi.org/10.3390/ijms26157441 (registering DOI) - 1 Aug 2025
Abstract
Gut microbiota-derived trimethylamine N-oxide (TMAO) has been implicated in both intestinal and renal diseases; however, its specific role in modulating gut–kidney interactions remains unclear. This study aimed to investigate the effects of TMAO on gut–kidney crosstalk using a mouse model of diarrhea. Mice [...] Read more.
Gut microbiota-derived trimethylamine N-oxide (TMAO) has been implicated in both intestinal and renal diseases; however, its specific role in modulating gut–kidney interactions remains unclear. This study aimed to investigate the effects of TMAO on gut–kidney crosstalk using a mouse model of diarrhea. Mice were divided into four groups: normal, model, TMAO, and TMAO + model. The normal group received sterile water, while the other groups were administered adenine + Folium sennae, TMAO, or a combination of TMAO and adenine + Folium sennae. Samples were collected to assess morphological changes in the colon and kidney, evaluate the colonic mucosal barrier and renal function, and measure NLRP3 inflammasome activity and inflammatory cytokine levels in colonic and renal tissues. TMAO levels and the gut microbiota composition were analyzed using 16S rRNA sequencing. The model group exhibited altered stool morphology, which was further aggravated by TMAO intervention. Both the model and TMAO + model groups exhibited significant damage to intestinal and renal tissues, along with compromised intestinal mucosal barriers and impaired renal function compared to controls. Inflammatory markers were elevated in these groups, with the TMAO + model group showing the most pronounced increases. Correlation analysis indicated significant relationships among TMAO levels, inflammasome activation, and inflammatory cytokines. The genera Mucispirillum and Anaerotruncus negatively correlated with TMAO, whereas Parabacteroides and Parasutterella genera positively correlated with TMAO. In conclusion, TMAO plays a critical role in modulating gut–kidney crosstalk by promoting inflammation, disrupting mucosal and renal integrity, and altering the gut microbial ecosystem. Full article
(This article belongs to the Collection Advances in Cell and Molecular Biology)
Show Figures

Figure 1

16 pages, 1868 KiB  
Article
Oral Candidiasis Associated with Aging and Salivary Hypofunction in Stomatitis Patients
by Yeon-Hee Lee, Solsol Seo, Tae-Seok Kim and Sang-woo Lee
J. Fungi 2025, 11(8), 574; https://doi.org/10.3390/jof11080574 (registering DOI) - 1 Aug 2025
Abstract
Objectives: Stomatitis is a broad term for oral mucosal inflammation, and oral candidiasis represents one of its common subtypes caused by fungal infection. This study aimed to investigate the relationship between oral candidiasis and reduced salivary flow in patients diagnosed with stomatitis and [...] Read more.
Objectives: Stomatitis is a broad term for oral mucosal inflammation, and oral candidiasis represents one of its common subtypes caused by fungal infection. This study aimed to investigate the relationship between oral candidiasis and reduced salivary flow in patients diagnosed with stomatitis and to identify clinical predictors of oral candidiasis. Methods: A total of 259 patients (mean age 59.77 ± 15.93 years; range 10–87 years; 201 females) with stomatitis were evaluated for oral candidiasis through Candida albicans culture testing. Clinical characteristics were compared between Candida-positive and Candida-negative groups. Unstimulated salivary flow rate (UFR) and stimulated salivary flow rate (SFR) were measured to assess xerostomia. Results: Among the 259 patients, 81 (31.3%) were diagnosed with oral candidiasis. Patients with candidiasis were significantly older (64.25 ± 14.66 years) than those without (57.73 ± 16.10 years; p = 0.002). Both UFR (0.36 ± 0.32 vs. 0.47 ± 0.28 mL/min, p = 0.006) and SFR (1.21 ± 0.68 vs. 1.41 ± 0.69 mL/min, p = 0.032) were significantly lower in the candidiasis group. The prevalence of xerostomia was significantly higher among Candida-positive patients, based on UFR ≤ 0.2 mL/min (49.4% vs. 18.5%, p < 0.001) and SFR ≤ 0.7 mL/min (27.2% vs. 10.7%, p < 0.001). The predictive accuracy for oral candidiasis was 62.2% based on age (AUC = 0.622; cutoff 64.50 years), 65.8% for UFR (AUC = 0.658; cutoff 0.335 mL/min), and 58.7% for SFR (AUC = 0.587; cutoff 1.150 mL/min). In the generalized linear model, xerostomia, as defined by UFR, was a significant predictor of oral candidiasis (B = 0.328, 95% CI: 0.177–0.480, p < 0.001). Conclusions: Oral candidiasis in patients with stomatitis was more strongly associated with decreased UFR than with aging alone. Among the factors assessed, reduced unstimulated salivary flow may serve as a useful clinical indicator for predicting oral candidiasis, particularly in elderly individuals. Full article
Show Figures

Figure 1

20 pages, 3054 KiB  
Article
Development of COVID-19 Vaccine Candidates Using Attenuated Recombinant Vesicular Stomatitis Virus Vectors with M Protein Mutations
by Mengqi Chang, Hui Huang, Mingxi Yue, Yuetong Jiang, Siping Yan, Yiyi Chen, Wenrong Wu, Yibing Gao, Mujin Fang, Quan Yuan, Hualong Xiong and Tianying Zhang
Viruses 2025, 17(8), 1062; https://doi.org/10.3390/v17081062 - 30 Jul 2025
Viewed by 211
Abstract
Recombinant vesicular stomatitis virus (rVSV) is a promising viral vaccine vector for addressing the COVID-19 pandemic. Inducing mucosal immunity via the intranasal route is an ideal strategy for rVSV-based vaccines, but it requires extremely stringent safety standards. In this study, we constructed two [...] Read more.
Recombinant vesicular stomatitis virus (rVSV) is a promising viral vaccine vector for addressing the COVID-19 pandemic. Inducing mucosal immunity via the intranasal route is an ideal strategy for rVSV-based vaccines, but it requires extremely stringent safety standards. In this study, we constructed two rVSV variants with amino acid mutations in their M protein: rVSV-M2 with M33A/M51R mutations and rVSV-M4 with M33A/M51R/V221F/S226R mutations, and developed COVID-19 vaccines based on these attenuated vectors. By comparing viral replication capacity, intranasal immunization, intracranial injection, and blood cell counts, we demonstrated that the M protein mutation variants exhibit significant attenuation effects both in vitro and in vivo. Moreover, preliminary investigations into the mechanisms of virus attenuation revealed that these attenuated viruses can induce a stronger type I interferon response while reducing inflammation compared to the wild-type rVSV. We developed three candidate vaccines against SARS-CoV-2 using the wildtype VSV backbone with either wild-type M (rVSV-JN.1) and two M mutant variants (rVSV-M2-JN.1 and rVSV-M4-JN.1). Our results confirmed that rVSV-M2-JN.1 and rVSV-M4-JN.1 retain strong immunogenicity while enhancing safety in hamsters. In summary, the rVSV variants with M protein mutations represent promising candidate vectors for mucosal vaccines and warrant further investigation. Full article
(This article belongs to the Special Issue Structure-Based Antiviral Drugs and Vaccine Design)
Show Figures

Figure 1

26 pages, 7326 KiB  
Article
Cocoa Polyphenols Alter the Fecal Microbiome Without Mitigating Colitis in Mice Fed Healthy or Western Basal Diets
by Eliza C. Stewart, Mohammed F. Almatani, Marcus Hayden, Giovanni Rompato, Jeremy Case, Samuel Rice, Korry J. Hintze and Abby D. Benninghoff
Nutrients 2025, 17(15), 2482; https://doi.org/10.3390/nu17152482 - 29 Jul 2025
Viewed by 195
Abstract
Background/Objectives: Chronic inflammation and Western-style diets elevate colorectal cancer (CRC) risk, particularly in individuals with colitis, a feature of inflammatory bowel disease (IBD). Diets rich in polyphenol-containing functional foods, such as cocoa, may reduce gut inflammation and modulate the gut microbiome. This [...] Read more.
Background/Objectives: Chronic inflammation and Western-style diets elevate colorectal cancer (CRC) risk, particularly in individuals with colitis, a feature of inflammatory bowel disease (IBD). Diets rich in polyphenol-containing functional foods, such as cocoa, may reduce gut inflammation and modulate the gut microbiome. This study investigated the impact of cocoa polyphenol (CP) supplementation on inflammation and microbiome composition in mice with colitis, fed either a healthy or Western diet, before, during, and after the onset of disease. We hypothesized that CPs would attenuate inflammation and promote distinct shifts in the microbiome, especially in the context of a Western diet. Methods: A 2 × 2 factorial design tested the effects of the basal diet (AIN93G vs. total Western diet [TWD]) and CP supplementation (2.6% w/w CocoaVia™ Cardio Health Powder). Inflammation was induced using the AOM/DSS model of colitis. Results: CP supplementation did not reduce the severity of colitis, as measured by disease activity index or histopathology. CPs did not alter gene expression in healthy tissue or suppress the colitis-associated pro-inflammatory transcriptional profile in either of the two diet groups. However, fecal microbiome composition shifted significantly with CPs before colitis induction, with persistent effects on several rare taxa during colitis and recovery. Conclusions: CP supplementation did not mitigate inflammation or mucosal injury at the tissue level, nor did it affect the expression of immune-related genes. While CPs altered microbiome composition, most notably in healthy mice before colitis, these shifts did not correspond to changes in inflammatory signaling. Basal diet remained the primary determinant of inflammation, mucosal damage, and colitis severity in this model. Full article
(This article belongs to the Section Phytochemicals and Human Health)
Show Figures

Figure 1

17 pages, 1899 KiB  
Article
Oat Fiber Alleviates Loperamide-Induced Constipation in Mice by Modulating Intestinal Barrier Function
by Yufei Shi, Yuchao Han, Jie Jiang, Di Wang, Zhongxia Li, Guiju Sun, Shaokang Wang, Wang Liao, Hui Xia, Da Pan and Ligang Yang
Nutrients 2025, 17(15), 2481; https://doi.org/10.3390/nu17152481 - 29 Jul 2025
Viewed by 189
Abstract
Objective: To investigate the effects of oat fiber on animal constipation and elucidate its underlying mechanisms. Methods: Male BALB/c mice were randomly allocated into five groups: control group (CON), model control group (MODEL), low dose group (LOW), middle dose group (MIDDLE), high dose [...] Read more.
Objective: To investigate the effects of oat fiber on animal constipation and elucidate its underlying mechanisms. Methods: Male BALB/c mice were randomly allocated into five groups: control group (CON), model control group (MODEL), low dose group (LOW), middle dose group (MIDDLE), high dose group (HIGH). Constipation was induced in the mice by intragastric administration of loperamide. Subsequently, the mice (except those in the CON and MODEL groups) were administered oat fiber intragastrically for 21 consecutive days. Results: Compared with the MODEL group, oat fiber significantly increased the number of fecal pellets, fecal wet weight, and fecal water content (p < 0.05), shortened the time to first black stool excretion (p < 0.05), and enhanced the small intestinal propulsion rate in constipated mice. Additionally, oat fiber significantly upregulated motilin (MTL) and gastrin (GAS) levels (p < 0.05), while downregulating vasoactive intestinal peptide (VIP) and somatostatin (SS) levels (p < 0.05). It also significantly reduced the transcription level of Aquaporin 8 (AQP8) (p < 0.05), effectively alleviating intestinal mucosal injury and immune inflammation. The relative expression levels of TNF-α and IL-1β were significantly decreased in the oat fiber group (p < 0.05). Gut microbiota analysis revealed that oat fiber increased both the abundance and diversity of gut microbiota in constipated mice. Specifically, oat fiber was found to enhance the relative abundance of Firmicutes while reducing that of Bacteroidetes. At the genus level, it promoted the proliferation of Lachnospiraceae_NK4A136_group and Roseburia. Conclusions: Oat fiber alleviates constipation in mice by modulating gastrointestinal regulatory peptides, gut microbiota, aquaporin and mitigating intestinal barrier damage and immune-inflammatory responses. Full article
(This article belongs to the Section Prebiotics and Probiotics)
Show Figures

Figure 1

19 pages, 4179 KiB  
Article
Camel Milk-Derived Extracellular Vesicles as a Functional Food Component Ameliorate Hypobaric Hypoxia-Induced Colonic Injury Through Microbiota–Metabolite Crosstalk
by Hui Yang, Demtu Er, Yu-Huan Wang, Bin-Tao Zhai and Rili Ge
Nutrients 2025, 17(15), 2431; https://doi.org/10.3390/nu17152431 - 25 Jul 2025
Viewed by 294
Abstract
Background/Objectives: This study investigates the therapeutic potential of camel milk-derived extracellular vesicles (CM-EVs) for treating colonic damage caused by high-altitude hypoxia, supporting the WHO’s “Food as Medicine” initiative. Methods: Using a 5500 m mouse model, researchers induced colonic injury and treated it with [...] Read more.
Background/Objectives: This study investigates the therapeutic potential of camel milk-derived extracellular vesicles (CM-EVs) for treating colonic damage caused by high-altitude hypoxia, supporting the WHO’s “Food as Medicine” initiative. Methods: Using a 5500 m mouse model, researchers induced colonic injury and treated it with oral CM-EVs for 15 days, comparing results to whole camel milk. Results: CM-EVs outperformed whole milk, significantly improving colon health by restoring barrier integrity and reducing disease activity index (DAI) (p < 0.01). They boosted beneficial bacteria like Lactobacillus and Bifidobacterium and decreased Enterobacteriaceae (p < 0.01). Metabolic analysis showed restored bile acid balance and amino acid modulation via the FXR/NF-κB pathway, reducing TLR4/MyD88-mediated inflammation and oxidative stress (p < 0.01). Fecal microbiota transplantation in the CM-EVs group notably decreased DAI and increased colon length (p < 0.05). Conclusions: CM-EVs repair mucosal damage, balance microbiota, and regulate metabolism to combat hypoxia-induced colonic damage, suggesting their potential as nutraceuticals and altitude-adaptive foods. This showcases nanotechnology’s role in enhancing traditional dietary benefits via precision nutrition. Full article
Show Figures

Figure 1

38 pages, 4533 KiB  
Review
A Narrative Review on the Multifaceted Roles of Galectins in Host–Pathogen Interactions During Helicobacter pylori Infection
by Bojan Stojanovic, Natasa Zdravkovic, Marko Petrovic, Ivan Jovanovic, Bojana S. Stojanovic, Milica Dimitrijevic Stojanovic, Jelena Nesic, Milan Paunovic, Ivana Milivojcevic Bevc, Nikola Mirkovic, Mladen Pavlovic, Nenad Zornic, Bojan Milosevic, Danijela Tasic-Uros, Jelena Zivic, Goran Colakovic and Aleksandar Cvetkovic
Int. J. Mol. Sci. 2025, 26(15), 7216; https://doi.org/10.3390/ijms26157216 - 25 Jul 2025
Viewed by 153
Abstract
Helicobacter pylori infection represents one of the most prevalent and persistent bacterial infections worldwide, closely linked to a spectrum of gastroduodenal diseases, including chronic gastritis, peptic ulceration, and gastric cancer. Recent advances have shed light on the critical role of endogenous lectins, particularly [...] Read more.
Helicobacter pylori infection represents one of the most prevalent and persistent bacterial infections worldwide, closely linked to a spectrum of gastroduodenal diseases, including chronic gastritis, peptic ulceration, and gastric cancer. Recent advances have shed light on the critical role of endogenous lectins, particularly galectins, in modulating host–pathogen interactions within the gastric mucosa. Galectins are β-galactoside-binding proteins with highly conserved structures but diverse biological functions, ranging from regulation of innate and adaptive immunity to modulation of cell signaling, apoptosis, and epithelial integrity. This review provides a comprehensive synthesis of current knowledge on the involvement of key galectin family members—especially Galectin-1, -2, -3, -8, and -9—in the context of H. pylori infection. Their dual roles in enhancing mucosal defense and facilitating bacterial persistence are examined along with their contributions to immune evasion, inflammation, and gastric carcinogenesis. Understanding the interplay between galectins and H. pylori enhances our knowledge of mucosal immunity. This interaction may also reveal potential biomarkers for disease progression and identify novel therapeutic targets. Modulating galectin-mediated pathways could improve outcomes in H. pylori-associated diseases. Full article
(This article belongs to the Special Issue New Insights into Lectins)
Show Figures

Figure 1

18 pages, 344 KiB  
Review
Intestinal Microbiota and Fecal Transplantation in Patients with Inflammatory Bowel Disease and Clostridioides difficile: An Updated Literature Review
by Chloe Lahoud, Toni Habib, Daniel Kalta, Reem Dimachkie, Suzanne El Sayegh and Liliane Deeb
J. Clin. Med. 2025, 14(15), 5260; https://doi.org/10.3390/jcm14155260 - 25 Jul 2025
Viewed by 377
Abstract
Background/Objectives: Inflammatory bowel disease (IBD) is characterized by chronic relapsing and remitting inflammation of the gastrointestinal tract. Fecal microbiota transplantation (FMT) has emerged as an FDA-approved treatment for recurrent Clostridioides difficile infections (CDIs), with promising potential in patients with IBD. This manuscript [...] Read more.
Background/Objectives: Inflammatory bowel disease (IBD) is characterized by chronic relapsing and remitting inflammation of the gastrointestinal tract. Fecal microbiota transplantation (FMT) has emerged as an FDA-approved treatment for recurrent Clostridioides difficile infections (CDIs), with promising potential in patients with IBD. This manuscript aimed to provide a comprehensive and updated review of the available literature on fecal microbiota transplantation, its clinical use in IBD in general, as well as in patients with IBD and CDI. Methods: An extensive literature search was performed from October 2024 to March 2025. All publications available within PubMed, Medline, Embase, Google Scholar, and Cochrane databases were reviewed. All original articles, case reports, review articles, systematic reviews, and meta-analyses were included. Qualitative and quantitative data were both extracted. Discussion: Intestinal microbiota is an integral part of the human body, and dysbiosis (an imbalance in the gut’s microbial community) has been linked with several pathologies. Dysbiosis in IBD is marked by reduced beneficial bacteria and increased pro-inflammatory pathogens, contributing to mucosal damage and immune dysregulation. FMT has emerged as a solution to dysbiosis, with the first case recorded in 1917. FMT has been successful in treating patients with CDI. The diagnostic value of the gut microbiome is currently being explored as a possible therapeutic approach to IBD. Several studies have assessed FMT in patients with IBD and CDI with promising results in both ulcerative colitis (UC) and Crohn’s disease (CD) but varying efficacy based on administration routes, donor selection, and processing methods. In the context of recurrent CDI in patients with IBD, FMT demonstrates a high cure rate and potential benefit in concurrently improving IBD activity. However, risks such as IBD flare-ups post-FMT remain a concern. Conclusions: FMT holds promising potential in the management of CDI in patients with IBD. By restoring microbial diversity and correcting dysbiosis, FMT offers a novel, microbiota-targeted alternative to conventional therapies. While data support its efficacy in improving disease remission, variability in outcomes underscores the need for standardized protocols and additional large-scale, controlled studies. Continued research efforts into donor selection, treatment regimens, and long-term safety will be critical to optimizing FMT’s role in IBD and CDI care as well as improving patient outcomes. Full article
(This article belongs to the Special Issue Emerging Treatment Options in Inflammatory Bowel Disease)
28 pages, 2482 KiB  
Article
Characterization of microRNA Expression Profiles of Murine Female Genital Tracts Following Nippostrongylus brasiliensis and Herpes Simplex Virus Type 2 Co-Infection
by Roxanne Pillay, Pragalathan Naidoo and Zilungile L. Mkhize-Kwitshana
Microorganisms 2025, 13(8), 1734; https://doi.org/10.3390/microorganisms13081734 - 24 Jul 2025
Viewed by 371
Abstract
Soil-transmitted helminths (STHs) and Herpes Simplex Virus type 2 (HSV-2) are highly prevalent infections with overlapping distribution, particularly in resource-poor regions. STH/HSV-2 co-infections may impact female reproductive health. However, many aspects of STH/HSV-2 co-infections, including the role of microRNAs (miRNAs) in regulating female [...] Read more.
Soil-transmitted helminths (STHs) and Herpes Simplex Virus type 2 (HSV-2) are highly prevalent infections with overlapping distribution, particularly in resource-poor regions. STH/HSV-2 co-infections may impact female reproductive health. However, many aspects of STH/HSV-2 co-infections, including the role of microRNAs (miRNAs) in regulating female genital tract (FGT) immunity and their potential contribution to pathologies such as chronic inflammation, impaired mucosal defense, and reproductive tract cancers remain unclear. In this study we investigated the miRNA expression profiles in murine FGT tissues following single or co-infection with Nippostrongylus brasiliensis (Nb) and HSV-2 and explored predicted miRNA-mRNA targets and pathways. An analysis of miRNA sequencing data was conducted to determine differentially expressed (DE) miRNAs between infected FGT tissues and uninfected controls. Ingenuity Pathway Analysis was conducted to predict the immune-related target genes of the DE miRNAs and reveal enriched canonical pathways, top diseases, and biological functions. Selected representative DE miRNAs were validated using RT-qPCR. Our results showed a total of eight DE miRNAs (mmu-miR-218-5p, mmu-miR-449a-5p, mmu-miR-497a-3p, mmu-miR-144-3p, mmu-miR-33-5p, mmu-miR-451a, mmu-miR-194-5p, and mmu-miR-192-5p) in the comparison of Nb-infected versus uninfected controls; nine DE miRNAs (mmu-miR-451a, mmu-miR-449a-5p, mmu-miR-144-3p, mmu-miR-376a-3p, mmu-miR-192-5p, mmu-miR-218-5p, mmu-miR-205-3p, mmu-miR-103-3p, and mmu-miR-200b-3p) in the comparison of HSV-2-infected versus uninfected controls; and one DE miRNA (mmu-miR-199a-5p) in the comparison of Nb/HSV-2 co-infected versus uninfected controls (p-value < 0.05, |logFC| ≥ 1). Core expression analysis showed that, among other canonical pathways, the DE miRNAs and their predicted mRNA targets were involved in neutrophil degranulation, interleukin-4 and interleukin-13 signaling, natural killer cell signaling, interferon alpha/beta signaling, and ISGylation. Additionally, cancer was predicted as one of the significantly enriched diseases, particularly in the co-infected group. This is the first study to provide insights into the FGT miRNA profiles following Nb and HSV-2 single and co-infection, as well as the predicted genes and pathways they regulate, which may influence host immunity and pathology. This study highlights the role of miRNAs in regulating FGT immunity and pathology in the context of STH/HSV-2 co-infection. Full article
(This article belongs to the Special Issue Insights into Microbial Infections, Co-Infections, and Comorbidities)
Show Figures

Figure 1

35 pages, 2034 KiB  
Review
The Role of Gut Microbiota in Gastrointestinal Immune Homeostasis and Inflammation: Implications for Inflammatory Bowel Disease
by Elisabetta Bretto, Miquel Urpì-Ferreruela, Gherzon Rimer Casanova and Begoña González-Suárez
Biomedicines 2025, 13(8), 1807; https://doi.org/10.3390/biomedicines13081807 - 24 Jul 2025
Viewed by 575
Abstract
Inflammatory bowel disease (IBD), a heterogeneous group of recurring inflammatory conditions of the digestive system that encompass both ulcerative colitis (UC) and Crohn’s disease (CD), pose a significant public health challenge, currently lacking a definitive cure. The specific etiopathogenesis of IBD is not [...] Read more.
Inflammatory bowel disease (IBD), a heterogeneous group of recurring inflammatory conditions of the digestive system that encompass both ulcerative colitis (UC) and Crohn’s disease (CD), pose a significant public health challenge, currently lacking a definitive cure. The specific etiopathogenesis of IBD is not yet fully understood, but a multifactorial interplay of genetic and environmental factors is suspected. A growing body of evidence supports the involvement of intestinal dysbiosis in the development of IBD, including the effects of dysbiosis on the integrity of the intestinal epithelial barrier, modulation of the host immune system, alterations in the enteric nervous system, and the perpetuation of chronic inflammation. A comprehensive understanding of these mechanisms is important to define preventive measures, to develop new effective and lasting treatments, and to improve disease outcome. This review examines the complex tri-directional relationship between gut microbiota, mucosal immune system, and intestinal epithelium in IBD. In addition, nonpharmacological and behavioral strategies aimed at restoring a proper microbial–immune relationship will be suggested. Full article
Show Figures

Figure 1

27 pages, 4050 KiB  
Article
The Gut Mycobiome and Nutritional Status in Paediatric Phenylketonuria: A Cross-Sectional Pilot Study
by Malgorzata Ostrowska, Elwira Komoń-Janczara, Bozena Mikoluc, Katarzyna Iłowiecka, Justyna Jarczak, Justyna Zagórska, Paulina Zambrzycka, Silvia Turroni and Hubert Szczerba
Nutrients 2025, 17(15), 2405; https://doi.org/10.3390/nu17152405 - 23 Jul 2025
Viewed by 218
Abstract
Background: Phenylketonuria (PKU) is a metabolic disorder managed through a strict, lifelong low-phenylalanine diet, which may influence gut microbiome dynamics. While gut bacterial alterations in PKU are increasingly investigated, the fungal community (mycobiome) remains largely unexplored. This study compared gut mycobiome composition and [...] Read more.
Background: Phenylketonuria (PKU) is a metabolic disorder managed through a strict, lifelong low-phenylalanine diet, which may influence gut microbiome dynamics. While gut bacterial alterations in PKU are increasingly investigated, the fungal community (mycobiome) remains largely unexplored. This study compared gut mycobiome composition and dietary profiles of paediatric PKU patients and healthy controls, stratified by age (<10 and 10–18 years). Methods: Stool samples from 20 children (10 PKU, 10 controls) were analysed using ITS1/ITS2 amplicon sequencing. Nutritional status was assessed using Body Mass Index percentiles (Polish standards), and nutrient intake was evaluated from three-day dietary records compared to national reference values. Correlations between fungal taxa and dietary factors were explored. Results: Although alpha diversity did not differ significantly, beta diversity and LEfSe analyses revealed distinct fungal profiles between PKU patients and controls, indicating a trend toward group separation (PERMANOVA: F = 1.54646, p = 0.09; ANOVA: p = 0.0609). PKU patients showed increased Eurotiales (p = 0.029), Aspergillaceae (p = 0.029), and Penicillium (p = 0.11) and decreased Physalacriaceae (0% vs. 5.84% in controls) and Malassezia (p = 0.13). Spearman’s analysis showed significant correlations between Geotrichum and intake of protein (ρ = 0.55, p = 0.0127) and phenylalanine (ρ = 0.70, p = 0.0005). Conclusions: Dietary treatment in PKU is associated with age-dependent shifts in the gut mycobiome, notably increasing the abundance of taxa such as Eurotiales, Aspergillaceae, and Penicillium, involved in carbohydrate/lipid metabolism and mucosal inflammation. These findings highlight the potential of gut fungi as nutritional and clinical biomarkers in PKU. Full article
(This article belongs to the Special Issue Nutrients: 15th Anniversary)
Show Figures

Figure 1

37 pages, 4312 KiB  
Review
Neutrophils and NETs in Pathophysiology and Treatment of Inflammatory Bowel Disease
by Marina Ortega-Zapero, Raquel Gomez-Bris, Ines Pascual-Laguna, Angela Saez and Jose M. Gonzalez-Granado
Int. J. Mol. Sci. 2025, 26(15), 7098; https://doi.org/10.3390/ijms26157098 - 23 Jul 2025
Viewed by 422
Abstract
Inflammatory Bowel Disease (IBD), which includes ulcerative colitis (UC) and Crohn’s disease (CD), results from dysregulated immune responses that drive chronic intestinal inflammation. Neutrophils, as key effectors of the innate immune system, contribute to IBD through multiple mechanisms, including the release of reactive [...] Read more.
Inflammatory Bowel Disease (IBD), which includes ulcerative colitis (UC) and Crohn’s disease (CD), results from dysregulated immune responses that drive chronic intestinal inflammation. Neutrophils, as key effectors of the innate immune system, contribute to IBD through multiple mechanisms, including the release of reactive oxygen species (ROS), pro-inflammatory cytokines, and neutrophil extracellular traps (NETs). NETs are web-like structures composed of DNA, histones, and associated proteins including proteolytic enzymes and antimicrobial peptides. NET formation is increased in IBD and has a context-dependent role; under controlled conditions, NETs support antimicrobial defense and tissue repair, whereas excessive or dysregulated NETosis contributes to epithelial injury, barrier disruption, microbial imbalance, and thrombotic risk. This review examines the roles of neutrophils and NETs in IBD. We summarize recent single-cell and spatial-omics studies that reveal extensive neutrophil heterogeneity in the inflamed gut. We then address the dual role of neutrophils in promoting tissue damage—through cytokine release, immune cell recruitment, ROS production, and NET formation—and in supporting microbial clearance and mucosal healing. We also analyze the molecular mechanisms regulating NETosis, as well as the pathways involved in NET degradation and clearance. Focus is given to the ways in which NETs disrupt the epithelial barrier, remodel the extracellular matrix, contribute to thrombosis, and influence the gut microbiota. Finally, we discuss emerging therapeutic strategies aimed at restoring NET homeostasis—such as PAD4 inhibitors, NADPH oxidase and ROS pathway modulators, and DNase I—while emphasizing the need to preserve antimicrobial host defenses. Understanding neutrophil heterogeneity and NET-related functions may facilitate the development of new therapies and biomarkers for IBD, requiring improved detection tools and integrated multi-omics and clinical data. Full article
Show Figures

Figure 1

24 pages, 6281 KiB  
Article
Bioactive Polysaccharides Prevent Lipopolysaccharide-Induced Intestinal Inflammation via Immunomodulation, Antioxidant Activity, and Microbiota Regulation
by Mingyang Gao, Wanqing Zhang, Yan Ma, Tingting Liu, Sijia Wang, Shuaihu Chen, Zhengli Wang and Hong Shen
Foods 2025, 14(15), 2575; https://doi.org/10.3390/foods14152575 - 23 Jul 2025
Viewed by 306
Abstract
Intestinal inflammation involves barrier impairment, immune hyperactivation, and oxidative stress imbalance. Bioactive polysaccharides universally alleviate inflammation via anti-inflammatory, antioxidant, and microbiota-modulating effects, yet exhibit distinct core mechanisms. Elucidating these differences is vital for targeted polysaccharide applications. This research examines distinct regulatory pathways through [...] Read more.
Intestinal inflammation involves barrier impairment, immune hyperactivation, and oxidative stress imbalance. Bioactive polysaccharides universally alleviate inflammation via anti-inflammatory, antioxidant, and microbiota-modulating effects, yet exhibit distinct core mechanisms. Elucidating these differences is vital for targeted polysaccharide applications. This research examines distinct regulatory pathways through which diverse bioactive polysaccharides mitigate lipopolysaccharide-triggered intestinal inflammation in male Kunming (KM) mice. This experiment employed Lentinula edodes polysaccharide (LNT), Auricularia auricula polysaccharide (AAP), Cordyceps militaris polysaccharide (CMP), Lycium barbarum polysaccharide (LBP), and Brassica rapa polysaccharide (BRP). The expression levels of biomarkers associated with the TLR4 signaling pathway, oxidative stress, and intestinal barrier function were quantified, along with comprehensive gut microbiota profiling. The results showed that all five polysaccharides alleviated inflammatory responses in mice by inhibiting inflammatory cytokine release, reducing oxidative damage, and modulating gut microbiota, but their modes of action differed: LBP significantly suppressed the TLR-4/MyD88 signaling pathway and its downstream pro-inflammatory cytokine expression, thereby blocking inflammatory signal transduction and reducing oxidative damage; LNT and CMP enhanced the body’s antioxidant capacity by increasing antioxidant enzyme activities and decreasing malondialdehyde (MDA) levels; AAP and BRP enriched Akkermansia (Akk.) within the Verrucomicrobia (Ver.) phylum, upregulating tight junction protein expression to strengthen the intestinal mucosal barrier and indirectly reduce oxidative damage. This research demonstrates that different polysaccharides alleviate inflammation through multi-target synergistic mechanisms: LBP primarily inhibits inflammatory pathways; AAP and BRP focus on intestinal barrier protection and microbiota modulation; and LNT and CMP exert effects via antioxidant enzyme activation. These data support designing polysaccharide blends that leverage complementary inflammatory modulation mechanisms. Full article
Show Figures

Figure 1

17 pages, 1402 KiB  
Review
Rethinking Short-Chain Fatty Acids: A Closer Look at Propionate in Inflammation, Metabolism, and Mucosal Homeostasis
by Sonia Facchin, Matteo Calgaro and Edoardo V. Savarino
Cells 2025, 14(15), 1130; https://doi.org/10.3390/cells14151130 - 22 Jul 2025
Viewed by 285
Abstract
Propionate is a short-chain fatty acid (SCFA) produced by gut microbiota through the fermentation of dietary fibers. Among the SCFAs, butyrate stands out and has been extensively studied for its beneficial effects; however, propionate has received less attention despite its relevant roles in [...] Read more.
Propionate is a short-chain fatty acid (SCFA) produced by gut microbiota through the fermentation of dietary fibers. Among the SCFAs, butyrate stands out and has been extensively studied for its beneficial effects; however, propionate has received less attention despite its relevant roles in immune modulation, metabolism, and mucosal homeostasis. This narrative review focuses on propionate’s effects on metabolism, inflammation, microbiota, and gastrointestinal diseases. Propionate acts as a signalling molecule through FFAR2/FFAR3 receptors and modulates immunity, energy metabolism, and gut–brain communication. It has beneficial effects in metabolic disorders, inflammatory bowel disease (IBD), and alcohol-related liver disease (ALD). However, excessive accumulation is linked to neurotoxicity, autism spectrum disorder (ASD), and mitochondrial dysfunction. Its effects are dose-dependent and tissue-specific, with both protective and harmful potentials depending on the context. Propionate use requires a personalized approach, considering the pathological context, host microbiota composition, and appropriate dosage to avoid adverse effects. Full article
Show Figures

Graphical abstract

15 pages, 7497 KiB  
Article
Hydrogel-Shielded Ellagic Acid Nanoparticles Prolong Colonic Retention and Mitigate DSS-Induced Colitis via Reactive Oxygen Species Scavenging
by Ximei Ye, Tao Chen, Lihang Chen, Di Wu, Yinan Du and Jiangning Hu
Foods 2025, 14(15), 2559; https://doi.org/10.3390/foods14152559 - 22 Jul 2025
Viewed by 238
Abstract
Inflammatory bowel disease (IBD) is characterized by oxidative stress imbalance and intestinal barrier disruption. Reducing excessive ROS has become a promising therapeutic strategy. Compared with conventional polyphenols, nanomaterials offer greater stability and bioavailability for ROS scavenging. Here, ellagic acid (EA) was converted into [...] Read more.
Inflammatory bowel disease (IBD) is characterized by oxidative stress imbalance and intestinal barrier disruption. Reducing excessive ROS has become a promising therapeutic strategy. Compared with conventional polyphenols, nanomaterials offer greater stability and bioavailability for ROS scavenging. Here, ellagic acid (EA) was converted into uniform nanoparticles (EAs) with reactive oxygen scavenging capacity through horseradish peroxidase (HRP)-mediated oxidative polymerization and subsequently encapsulated in the anti-gastric acid hydrogel F-DP to obtain the hybrid system F-DP@EAs. EAs reduced ROS, MDA, NO, IL-1β, and TNF-α levels in vitro, while increasing IL-4 and IL-10 expression, thus alleviating inflammation. Herein, F-DP@EAs prolonged intestinal retention time and exerted superior protective effects in the DSS-induced colitis model. Oral F-DP@EAs lowered DAI, preserved colon length, increased glutathione (GSH) and superoxide dismutase (SOD), decreased NO and MDA, restored zonula occludens-1 (ZO-1), and reduced mucosal lesions. These findings demonstrate that combining nanoparticle and hydrogel technologies markedly enhances the preventive and protective efficacy of EA, highlighting F-DP@EAs as a promising candidate for future IBD therapy. Full article
(This article belongs to the Section Food Nutrition)
Show Figures

Figure 1

Back to TopTop